1
|
Liu Y, Li D, Zhang T, Wang K, Liang X, Zong X, Yang H, Li Z. Effect of imatinib on lipopolysaccharide‑induced acute lung injury and endothelial dysfunction through the P38 MAPK and NF-κB signaling pathways in vivo and in vitro. Respir Physiol Neurobiol 2024; 333:104388. [PMID: 39725368 DOI: 10.1016/j.resp.2024.104388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND The primary purpose of this study was to demonstrate the preventive effects of imatinib (IMA) on lipopolysaccharide (LPS)-induced inflammation in a mouse model of acute lung injury (ALI) and human umbilical vascular endothelial cells. METHODS LPS stimulation for 24 h induced ALI and cell inflammation. The pathological results of the lungs were evaluated using the wet/dry weight ratio, pulmonary vascular permeability measurements, and myeloperoxidase immunohistochemistry. The expression of pro-inflammatory mediators was analyzed using RT-PCR and enzyme-linked immunosorbent assay. Protein levels were analyzed using western blotting. The structure of cell junctions was detected using immunofluorescence. RESULTS IMA improved LPS-induced pulmonary pathological damage and reduced the lung wet/dry weight ratio and myeloperoxidase expression in the lung tissue. IMA decreased bronchoalveolar lavage fluid inflammatory cell count and the release of tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and monocyte chemotactic protein 1 (MCP-1) in the blood. Pretreatment of human umbilical vascular endothelial cells with IMA significantly attenuated LPS-induced actin stress fiber formation and vascular endothelial-cadherin disruption. In addition, IMA downregulated the mRNA abundances of vascular cell adhesion molecule 1, intercellular adhesion molecule 1, IL-1β, IL-6, and tumor necrosis factor-α(TNF-α) expression. The phosphorylation of p65, nuclear factor-kappa B inhibitor alpha (IκBα), p38, extracellular signal-regulated kinase, and Jun N-terminal kinase induced by LPS were attenuated after IMA treatment in vivo and in vitro. CONCLUSIONS IMA modulates the nuclear factor-kappa B and mitogen-activated protein kinase signaling pathways and the production of pro-inflammatory cytokines to prevent cellular damage due to LPS infection. These results indicate that IMA may be a potential modulator of LPS-induced ALI.
Collapse
Affiliation(s)
- Yaru Liu
- Department of Emergency Medicine,The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Duanyang Li
- Department of Emergency Medicine,The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Tianyi Zhang
- Department of Emergency Medicine,The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Keruo Wang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211 China
| | - Xue Liang
- Tianjin Institute of Cardiology, Tianjin 300211,China
| | - Xiaolong Zong
- Department of Clinical Laboratory, The Second Hospital of Tianjin Medical University, Tianjin 300211,China
| | - Hong Yang
- School of Biomedical Engineering and The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China.
| | - Zhenyu Li
- Department of Emergency Medicine,The Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| |
Collapse
|
2
|
Younis NS, Almostafa MM, Mohamed ME. Geraniol Ameliorates Pentylenetetrazol-Induced Epilepsy, Neuroinflammation, and Oxidative Stress via Modulating the GABAergic Tract: In vitro and in vivo studies. Drug Des Devel Ther 2024; 18:5655-5672. [PMID: 39654600 PMCID: PMC11627104 DOI: 10.2147/dddt.s481985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 11/27/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction Geraniol (Ger), a monoterpene, is a common constituent of several essential oils. This study explored the anticonvulsant effect of Ger in-vitro using nerve growth factor (NGF) prompted PC12 cell injured by Glutamate (Glu) and in-vivo using Pentylenetetrazole (PTZ)-induced kindling through the GABAergic pathway. Materials To assess the effect of Ger on NGF prompted PC12 cells injured by Glu, Ger at concentrations of 25, 50, 100, 200 and 400 μg/mL was used. GABA, 5-HT, IL-1β, IL-4, and TNF-α levels and the gene expressions of GABAA-Rα1, NMDAR1, GAD 65, GAD 67, GAT 1 and GAT 3 were measured in NGF-induced PC12 cells treated with Ger (100, and 200 μg/mL). Mice were randomly separated into five groups. Normal and PTZ groups in which mice were injected with saline or PTZ, respectively. PTZ + Ger 100, PTZ + Ger 200 and PTZ + SV groups in which mice orally administered Ger or sodium valproate (SV), respectively, then injected with PTZ. Results Ger up to 400 μg/mL did not display any toxicity or injury in PC12 cells. Ger (100 to 200 μg/mL) reduced the injury induced by Glu, increased the gene expression of GABAA-Rα1, GAD65 and GAD67 and decreased GAT 1, GAT 3 and NMDAR1 expression in NGF-induced PC12 cells damaged by Glu. Ger (100 to 200 μg/mL) increased GABA and reduced TNF-α, IL-4 and IL-1β levels in NGF-induced PC12 cells injured by Glu. As for the in-vivo results, Ger increased GABA, GAD, GAT 1 and 3 and lowered GABA T. Ger mitigated MDA, NO, IL-1β, IL-6, TNF-α and IFN-γ, GFAP, caspase-3, and -9 levels and Bax gene expression and escalated GSH, SOD, catalase, BDNF and Bcl2 gene expression. Conclusion Ger reduced the oxidative stress status, neuroinflammation and apoptosis and activated GABAergic neurotransmission, which might clarify its anticonvulsant. Ger protects animals against PTZ prompted kindling as established by the enhancement in short term as well as long-term memory. Ger mitigated the injury induced by Glu in NGF prompted PC12 cell.
Collapse
Affiliation(s)
- Nancy S Younis
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Alhofuf, Al-Ahsa, 31982, Saudi Arabia
- Zagazig University Hospitals, Zagazig University, Zagazig, 44519, Egypt
| | - Mervt M Almostafa
- Department of Chemistry, College of Science, King Faisal University, Alhofuf, Al-Ahsa, 31982, Saudi Arabia
| | - Maged E Mohamed
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Alhofuf, Al-Ahsa, 31982, Saudi Arabia
- Pharmacognosy, College of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
3
|
Jiang K, Chen Y, Wang K, Yang L, Sun S, Yang J, Li X. miR-331-depleted exosomes derived from injured endometrial epithelial cells promote macrophage activation during endometritis. Int J Biol Macromol 2024; 279:134967. [PMID: 39179075 DOI: 10.1016/j.ijbiomac.2024.134967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Exosomes are natural carriers of biological macromolecules that are involved in the pathogenesis of a wide variety of inflammatory diseases. The purpose of this study was to investigate the role of exosomes derived from injured endometrial epithelial cells (EECs) in the development of endometritis. We isolated exosomes derived from LPS-injured EECs and identified these exosomes as proinflammatory mediators that can be internalized by macrophages and thus induce proinflammatory macrophage activation. We further found that miR-331 expression was sharply downregulated in exosomes derived from LPS-injured EECs and that macrophages treated with these exosomes also presented a lower level of miR-331. Importantly, the pathogenic role of exosomal miR-331 in promoting endometrial inflammation was revealed by the ability of adoptively transferred EECs-derived exosomes to cause macrophage activation, and this was reversed by miR-331 overexpression. Mechanistically, overexpression of miR-331 in macrophages mitigated NF-κB p65 phosphorylation by inhibiting the Notch1/IKKα pathway, which in turn curbed macrophage activation. In vivo assays further unveiled that miR-331 expression is negatively correlated with proinflammatory macrophage activation and that miR-331 upregulation markedly slowed disease progression in mice with endometritis. The exosome/miR-331/Notch1 axis plays a critical pathological role in endometrial inflammation, representing a new therapeutic target for endometritis.
Collapse
Affiliation(s)
- Kangfeng Jiang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Yajing Chen
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Kui Wang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Liangyu Yang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Shumin Sun
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, Yunnan, China.
| | - Jing Yang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, Yunnan, China.
| | - Xiaobing Li
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, Yunnan, China.
| |
Collapse
|
4
|
Li L, Song QQ, Li SR, Jia ZG, Sun XC, Zhao YT, Deng JB, Wu JJ, Ni T, Liu JS. Human umbilical cord mesenchymal stem cells-derived exosomes attenuate burn-induced acute lung injury via inhibiting ferroptosis. Acta Histochem 2024; 126:152189. [PMID: 39197328 DOI: 10.1016/j.acthis.2024.152189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024]
Abstract
Our previous study has shown that exosomes derived from human umbilical cord mesenchymal stem cells (hUCMSCs-exo) alleviated burn-induced acute lung injury (ALI). In this study, we explored a novel mechanism by which hUCMSCs-exo contributed to the inhibition of burn-induced ALI. The ALI rat model with severe burn was established for the in vivo experiments, and rats PMVECs were stimulated with the serum from burn-induced ALI rats for the in vitro experiments. The pathological changes of lung tissues were evaluated by HE staining; the cell viability was measured using CCK-8; the iron level and Fe2+ concentration were assessed using Iron Assay Kit and Fe2+ fluorescence detection probe; the mRNA expression of SLC7A11 and GPX4 were measured by qRT-PCR; the protein levels of SLC7A11, GPX4, Nrf2 and HO-1 were detected by western blot. Both the in vivo and in vitro experiments revealed that ferroptosis was significantly induced in burn-induced ALI, which as verified by increased iron level and Fe2+ concentration, and decreased SLC7A11 and GPX4 mRNA and protein levels. Furthermore, both hUCMSCs-exo and Fer-1 (the inhibitor of ferroptosis) alleviated lung inflammation and up-regulated protein levels of Nrf2 and HO-1 in the lung tissues of burn-induced ALI rats. These results suggested that hUCMSCs-exo exhibited a protective role against burn-induced ALI by inhibiting ferroptosis, partly owing to the activation of Nrf2/HO-1 pathway, thus providing a novel therapeutic strategy for burn-induced ALI.
Collapse
Affiliation(s)
- Lin Li
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China
| | - Qin-Qin Song
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China
| | - Shuang-Ru Li
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China
| | - Zhi-Gang Jia
- Department of Burn and Plastic Surgery, Affiliated Hospital of Jiangnan University, Wuxi 214028, China
| | - Xing-Chen Sun
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China
| | - Yu-Ting Zhao
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China
| | - Jia-Bin Deng
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China
| | - Jun-Jun Wu
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China
| | - Tao Ni
- Department of Burn and Plastic Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201900, China
| | - Ji-Song Liu
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China.
| |
Collapse
|
5
|
Zhang W, Wang X, Ma C, Liang B, Ma L, Wang Y, Lin Y, Han S. Pyroptosis inhibition alleviates acute lung injury via E-twenty-six variant gene 5-mediated downregulation of gasdermin D. Respir Physiol Neurobiol 2024; 331:104346. [PMID: 39265817 DOI: 10.1016/j.resp.2024.104346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/19/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND Acute lung injury (ALI) is a life-threatening condition characterized by excessive pulmonary inflammation, yet its precise pathophysiology remains elusive. Pyroptosis, a programmed cell death mechanism controlled by gasdermin D (GSDMD), has been linked to the etiology of ALI. This study investigated the regulatory functions of the transcription factor E-twenty-six variant gene 5 (ETV5) and GSDMD in ALI. METHODS Lipopolysaccharide (LPS) was used to treat BEAS-2B cells (50 mmol/mL) and establish an LPS-induced mouse model of ALI (by intratracheal administration, 3 mg/kg). Protein-protein docking, immunofluorescence analysis, western blotting, real-time quantitative polymerase chain reaction, and dual-luciferase reporter gene assay were used to examine ETV5-mediated negative feedback regulation of GSDMD and its effects on pyroptosis and ALI. RESULTS Our results showed that the physiological function of ETV5 was reduced by its downregulated expression, which impeded its nuclear translocation in ALI mice. Increased pyroptosis and enhanced production of inflammatory cytokines were associated with LPS-induced ALI. ETV5 overexpression in LPS-treated BEAS-2B cells decreased the expression of total and membrane-bound GSDMD, negatively regulated GSDMD, and prevented pyroptosis. The expression of inflammatory cytokines was subsequently reduced due to this inhibition, which, in turn, reduced ALI. Molecular docking analysis and dual-luciferase reporter gene assay results indicated a direct interaction between ETV5 and GSDMD, which inhibited GSDMD production. CONCLUSION Our results indicate that ETV5 inhibits pyroptosis, decreases the expression of inflammatory cytokines, and negatively regulates GSDMD expression to ameliorate ALI symptoms.
Collapse
Affiliation(s)
- Wenlong Zhang
- Department of Respiratory and Critical Care Medicine, Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, No. 68 Zhongshan Road, Wuxi City, Jiangsu Province 214000, China
| | - Xinhua Wang
- Department of Respiratory and Critical Care Medicine, Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, No. 68 Zhongshan Road, Wuxi City, Jiangsu Province 214000, China
| | - Chenhui Ma
- Department of Respiratory and Critical Care Medicine, Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, No. 68 Zhongshan Road, Wuxi City, Jiangsu Province 214000, China
| | - Bao Liang
- Department of Respiratory and Critical Care Medicine, Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, No. 68 Zhongshan Road, Wuxi City, Jiangsu Province 214000, China
| | - Lihong Ma
- Department of Respiratory and Critical Care Medicine, Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, No. 68 Zhongshan Road, Wuxi City, Jiangsu Province 214000, China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, No. 68 Zhongshan Road, Wuxi City, Jiangsu Province 214000, China
| | - Yuanjie Lin
- Department of Respiratory and Critical Care Medicine, Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, No. 68 Zhongshan Road, Wuxi City, Jiangsu Province 214000, China
| | - Shuguang Han
- Department of Respiratory and Critical Care Medicine, Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, No. 68 Zhongshan Road, Wuxi City, Jiangsu Province 214000, China.
| |
Collapse
|
6
|
Zhu X, Hua E, Tu Q, Liu M, Xu L, Feng J. Foxq1 Promotes Alveolar Epithelial Cell Death through Tle1-mediated Inhibition of the NF-κB Signaling Pathway. Am J Respir Cell Mol Biol 2024; 71:53-65. [PMID: 38574238 DOI: 10.1165/rcmb.2023-0317oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/03/2024] [Indexed: 04/06/2024] Open
Abstract
Acute lung injury (ALI) is a common respiratory disease characterized by diffuse alveolar injury and interstitial edema, as well as a hyperinflammatory response, lung cell damage, and oxidative stress. Foxq1, a member of the FOX family of transcription factors, is expressed in various tissues, such as the lungs, liver, and kidneys, and contributes to various biological processes, such as stress, metabolism, cell cycle arrest, and aging-related apoptosis. However, the role of Foxq1 in ALI is unknown. We constructed ex vivo and in vivo ALI models by LPS tracheal perfusion of ICR mice and conditioned medium stimulation of injured MLE-12 cells. Foxq1 expression was increased, and its localization was altered, in our ALI model. In normal or injured MLE-12 cells, knockdown of Foxq1 promoted cell survival, and overexpression had the opposite effect. This regulatory effect was likely mediated by Tle1 and the NF-κB/Bcl2/Bax signaling pathway. These data suggest a potential link between Foxq1 and ALI, indicating that Foxq1 can be used as a biomarker for the diagnosis of ALI. Targeted inhibition of Foxq1 expression could promote alveolar epithelial cell survival and may provide a strategy for mitigating ALI.
Collapse
Affiliation(s)
- Xi Zhu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital and Medical School of Nantong University, Nantong, China; and
| | - Ershi Hua
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital and Medical School of Nantong University, Nantong, China; and
| | - Qifeng Tu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Liqin Xu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital and Medical School of Nantong University, Nantong, China; and
| | - Jian Feng
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital and Medical School of Nantong University, Nantong, China; and
| |
Collapse
|
7
|
Li X, Zhang X, Kang Y, Cai M, Yan J, Zang C, Gao Y, Qi Y. Scutellarein Suppresses the Production of ROS and Inflammatory Mediators of LPS-Activated Bronchial Epithelial Cells and Attenuates Acute Lung Injury in Mice. Antioxidants (Basel) 2024; 13:710. [PMID: 38929149 PMCID: PMC11200809 DOI: 10.3390/antiox13060710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/06/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Scutellarein is a key active constituent present in many plants, especially in Scutellaria baicalensis Georgi and Erigeron breviscapus (vant.) Hand-Mazz which possesses both anti-inflammatory and anti-oxidative activities. It also is the metabolite of scutellarin, with the ability to relieve LPS-induced acute lung injury (ALI), strongly suggesting that scutellarein could suppress respiratory inflammation. The present study aimed to investigate the effects of scutellarein on lung inflammation by using LPS-activated BEAS-2B cells (a human bronchial epithelial cell line) and LPS-induced ALI mice. The results showed that scutellarein could reduce intracellular reactive oxygen species (ROS) accumulation through inhibiting the activation of NADPH oxidases, markedly downregulating the transcription and translation of pro-inflammatory cytokines, including interleukin-6 (IL-6), C-C motif chemokine ligand 2 (CCL2), and C-X-C motif chemokine ligand (CXCL) 8 in LPS-activated BEAS-2B cells. The mechanism study revealed that it suppressed the phosphorylation and degradation of IκBα, consequently hindering the translocation of p65 from the cytoplasm to the nucleus and its subsequent binding to DNA, thereby decreasing NF-κB-regulated gene transcription. Notably, scutellarein had no impact on the activation of AP-1 signaling. In LPS-induced ALI mice, scutellarein significantly decreased IL-6, CCL2, and tumor necrosis factor-α (TNF-α) levels in the bronchoalveolar lavage fluid, attenuated lung injury, and inhibited neutrophil infiltration. Our findings suggest that scutellarein may be a beneficial agent for the treatment of infectious pneumonia by virtue of its anti-oxidative and anti-inflammatory activities.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuan Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; (X.L.); (X.Z.); (Y.K.); (M.C.); (J.Y.); (C.Z.)
| | - Yun Qi
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; (X.L.); (X.Z.); (Y.K.); (M.C.); (J.Y.); (C.Z.)
| |
Collapse
|
8
|
Zhan J, Chen J, Deng L, Lu Y, Luo L. Exploring the ferroptosis-related gene lipocalin 2 as a potential biomarker for sepsis-induced acute respiratory distress syndrome based on machine learning. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167101. [PMID: 38423372 DOI: 10.1016/j.bbadis.2024.167101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Sepsis is a major cause of mortality in patients, and ARDS is one of the most common outcomes. The pathophysiology of acute respiratory distress syndrome (ARDS) caused by sepsis is significantly impacted by genes related to ferroptosis. METHODS In this study, Weighted gene co-expression network analysis (WGCNA), protein-protein interaction (PPI) networks, functional enrichment analysis, and machine learning were employed to identify characterized genes and to construct receiver operating characteristic (ROC) curves. Additionally, DNA methylation levels were quantified and single-cell analysis was conducted. To validate the alterations in the expression of Lipocalin-2 (LCN2) and ferroptosis-related proteins in the in vitro model, Western blotting was carried out, and the changes in intracellular ROS and Fe2+ levels were detected. RESULTS A combination of eight machine learning algorithms, including RFE, LASSO, RandomForest, SVM-RFE, GBDT, Bagging, XGBoost, and Boruta, were used with a machine learning model to highlight the significance of LCN2 as a key gene in sepsis-induced ARDS. Analysis of immune cell infiltration showed a positive correlation between neutrophils and LCN2. In a cell model induced by LPS, it was found that Ferrostatin-1 (Fer-1), a ferroptosis inhibitor, was able to reverse the expression of LCN2. Knocking down LCN2 in BEAS-2B cells reversed the LPS-induced lipid peroxidation, Fe2+ levels, ACSL4, and GPX4 levels, indicating that LCN2, a ferroptosis-related gene (FRG), plays a crucial role in mediating ferroptosis. CONCLUSION Upon establishing an FRG model for individuals with sepsis-induced ARDS, we determined that LCN2 could be a dependable marker for predicting survival in these patients. This finding provides a basis for more accurate ARDS diagnosis and the exploration of innovative treatment options.
Collapse
Affiliation(s)
- Jiayi Zhan
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Junming Chen
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Liyan Deng
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Yining Lu
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, Guangdong, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, Guangdong, China.
| |
Collapse
|
9
|
Rajendran P, Al-Saeedi FJ, Ammar RB, Abdallah BM, Ali EM, Al Abdulsalam NK, Tejavat S, Althumairy D, Veeraraghavan VP, Alamer SA, Bekhet GM, Ahmed EA. Geraniol attenuates oxidative stress and neuroinflammation-mediated cognitive impairment in D galactose-induced mouse aging model. Aging (Albany NY) 2024; 16:5000-5026. [PMID: 38517361 PMCID: PMC11006477 DOI: 10.18632/aging.205677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/13/2024] [Indexed: 03/23/2024]
Abstract
D-galactose (D-gal) administration was proven to induce cognitive impairment and aging in rodents' models. Geraniol (GNL) belongs to the acyclic isoprenoid monoterpenes. GNL reduces inflammation by changing important signaling pathways and cytokines, and thus it is plausible to be used as a medicine for treating disorders linked to inflammation. Herein, we examined the therapeutic effects of GNL on D-gal-induced oxidative stress and neuroinflammation-mediated memory loss in mice. The study was conducted using six groups of mice (6 mice per group). The first group received normal saline, then D-gal (150 mg/wt) dissolved in normal saline solution (0.9%, w/v) was given orally for 9 weeks to the second group. In the III group, from the second week until the 10th week, mice were treated orally (without anesthesia) with D-gal (150 mg/kg body wt) and GNL weekly twice (40 mg/kg body wt) four hours later. Mice in Group IV were treated with GNL from the second week up until the end of the experiment. For comparison of young versus elderly mice, 4 month old (Group V) and 16-month-old (Group VI) control mice were used. We evaluated the changes in antioxidant levels, PI3K/Akt levels, and Nrf2 levels. We also examined how D-gal and GNL treated pathological aging changes. Administration of GNL induced a significant increase in spatial learning and memory with spontaneously altered behavior. Enhancing anti-oxidant and anti-inflammatory effects and activating PI3K/Akt were the mechanisms that mediated this effect. Further, GNL treatment upregulated Nrf2 and HO-1 to reduce oxidative stress and apoptosis. This was confirmed using 99mTc-HMPAO brain flow gamma bioassays. Thus, our data suggested GNL as a promising agent for treating neuroinflammation-induced cognitive impairment.
Collapse
Affiliation(s)
- Peramaiyan Rajendran
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai 600077, Tamil Nadu, India
| | - Fatma J. Al-Saeedi
- Department of Nuclear Medicine, College of Medicine, Kuwait University, Safat 13110, Kuwait
| | - Rebai Ben Ammar
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Laboratory of Aromatic and Medicinal Plants, Center of Biotechnology of Borj-Cedria, Technopole of Borj-Cedria PBOX 901, Hammam-Lif 2050, Tunisia
| | - Basem M. Abdallah
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Enas M. Ali
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Najla Khaled Al Abdulsalam
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Sujatha Tejavat
- Department of Biomedical Sciences, College of Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Duaa Althumairy
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai 600077, Tamil Nadu, India
| | - Sarah Abdulaziz Alamer
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Gamal M. Bekhet
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Department of Zoology, Faculty of Science, Alexandria University Egypt, Alexandria 21544, Egypt
| | - Emad A. Ahmed
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Laboratory of Molecular Physiology, Zoology Department, Faculty of Science, Assiut University, Assiut 71515, Egypt
| |
Collapse
|
10
|
Nabil-Adam A, Ashour ML, Shreadah MA. Modulation of MAPK/NF-κB Pathway and NLRP3 Inflammasome by Secondary Metabolites from Red Algae: A Mechanistic Study. ACS OMEGA 2023; 8:37971-37990. [PMID: 37867644 PMCID: PMC10586274 DOI: 10.1021/acsomega.3c03480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/21/2023] [Indexed: 10/24/2023]
Abstract
The pharmacological properties of seaweeds are diverse. No studies have been conducted on the protective effect of Galaxaura oblongata (GOE) against lippopolysaccharide (LPS)-induced inflammation in the brain. This study is divided into three phases, the first of which is the initial phase. In vitro study includes antioxidant, radical scavenging, and anti-inflammatory activities, including cyclooxygenase-1 (COX1), COX2, NO, acetylcholine inhibition, sphingosine kinase 1, tumor necrosis factor α (TNF-α), and interleukin-6, as well as antioxidant and radical-scavenging activities, including 2,2-diphenyl-1-picrylhydrazyl and 2,2'-azinobis(3-ethylbenzothiazoline)-6-sulfonic acid. Using LPS-induced acute inflammation, the second phase was conducted in vivo. Antioxidant and anti-inflammatory assays were performed to investigate the protective role of GOE. In addition to the phytochemical analysis, the bioactive content of GOE was also investigated. In vitro results demonstrated the potential of GOE as an antioxidant, anti-inflammatory, and neuroprotective agent. A study using LPS as an induced lung injury and neuroinflammation model confirmed the in vitro results. The GOE significantly reduced inflammatory, oxidative, and neurodegenerative biomarkers based on histopathological and immuno-histochemistry results. Based on computational drug design, four target proteins were approved: nuclear factor κB, mitogen-activated protein kinases, TNF-α, and NLRP3. Using polyphenolic compounds in GOE as ligands demonstrated good alignment and affinity against the three proteins. Finally, the current study offers a new approach to developing drug leads considering GOE's protective and curative roles.
Collapse
Affiliation(s)
- Asmaa Nabil-Adam
- Marine
Biotechnology and Natural Products Laboratory, National Institute of Oceanography & Fisheries, Alexandria 21556, Egypt
| | - Mohamed L. Ashour
- Department
of Pharmacognosy, Faculty of Pharmacy, Ain-Shams
University, Abbasia, Cairo 11566, Egypt
- Department
of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P. O. Box
6231, Jeddah 21442, Saudi Arabia
| | - Mohamed Attia Shreadah
- Marine
Biotechnology and Natural Products Laboratory, National Institute of Oceanography & Fisheries, Alexandria 21556, Egypt
| |
Collapse
|
11
|
Malik MNH, Tahir MN, Alsahli TG, Tusher MMH, Alzarea SI, Alsuwayt B, Jahan S, Gomaa HAM, Shaker ME, Ali M, Anjum I, Khan MT, Roman M, Shabbir R. Geraniol Suppresses Oxidative Stress, Inflammation, and Interstitial Collagenase to Protect against Inflammatory Arthritis. ACS OMEGA 2023; 8:37128-37139. [PMID: 37841186 PMCID: PMC10568708 DOI: 10.1021/acsomega.3c04684] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023]
Abstract
Geraniol (GER) is a plant-derived acyclic isoprenoid monoterpene that has displayed anti-inflammatory effects in numerous in vivo and in vitro models. This study was therefore designed to evaluate the antiarthritic potential of GER in complete Freund's adjuvant (CFA)-induced inflammatory arthritis (IA) model in rats. IA was induced by intraplantar injection of CFA (0.1 mL), and a week after CFA administration, rats were treated with various doses of methotrexate (MTX; 1 mg/kg) or GER (25, 50, and 100 mg/kg). Treatments were given on every alternate day, and animals were sacrificed on the 35th day. Paw volume, histopathological, hematological, radiographic, and qPCR analyses were performed to analyze the severity of the disease. GER significantly reduced paw edema after 35 days of treatment, and these results were comparable to the MTX-treated group. GER-treated animals displayed a perfect joint structure with minimal inflammation and no signs of cartilage or bone damage. Moreover, GER restored red blood cell and hemoglobin levels, normalized erythrocyte sedimentation rate, platelet, and c-reactive protein values, and also attenuated the levels of rheumatoid factor. RT-qPCR analysis demonstrated that GER decreased mRNA expression of pro-inflammatory cytokines like tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta. GER also down-regulated the transcript levels of cyclooxygenase-2 (COX-2), microsomal prostaglandin E synthase-1, prostaglandin D2 synthase, and interstitial collagenase (MMP-1). Molecular docking of GER with COX-2, TNF-α, and MMP-1 also revealed that the antiarthritic effects of GER could be due to its direct interactions with these mediators. Based on our findings, it is conceivable that the antiarthritic effects of GER could be attributed to downregulation of pro-inflammatory mediators and protease like MMP-1.
Collapse
Affiliation(s)
- Muhammad Nasir Hayat Malik
- Faculty
of Pharmacy, Capital University of Science
and Technology (CUST), Islamabad 44000, Pakistan
- Faculty
of Pharmacy, The University of Lahore, Lahore 54000, Pakistan
| | | | - Tariq G. Alsahli
- Department
of Pharmacology, College of Pharmacy, Jouf
University, Sakaka, Aljouf 72341, Saudi Arabia
| | - Md. Mahedi Hassan Tusher
- Department
of Pharmacology, Faculty of Basic Sciences, Bangladesh University of Health Sciences, Dhaka 1216, Bangladesh
| | - Sami I. Alzarea
- Department
of Pharmacology, College of Pharmacy, Jouf
University, Sakaka, Aljouf 72341, Saudi Arabia
| | - Bader Alsuwayt
- Department
of Pharmacy Practice, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin 31991, Saudi Arabia
| | - Shah Jahan
- Department
of Immunology, University of Health Sciences, Lahore 54000, Pakistan
| | - Hesham A. M. Gomaa
- Department
of Pharmacology, College of Pharmacy, Jouf
University, Sakaka, Aljouf 72341, Saudi Arabia
| | - Mohamed E. Shaker
- Department
of Pharmacology, College of Pharmacy, Jouf
University, Sakaka, Aljouf 72341, Saudi Arabia
| | - Muhammad Ali
- Faculty
of Pharmacy, The University of Lahore, Lahore 54000, Pakistan
| | - Irfan Anjum
- Faculty
of Pharmacy, The University of Lahore, Lahore 54000, Pakistan
- Shifa
College of Pharmaceutical Sciences,Shifa
Tameer-e-Millat University, Islamabad 44000, Pakistan
| | - Muhammad Tariq Khan
- Faculty
of Pharmacy, Capital University of Science
and Technology (CUST), Islamabad 44000, Pakistan
| | - Muhammad Roman
- Department
of Microbiology, University of Health Sciences, Lahore 54000, Pakistan
| | - Ramla Shabbir
- Faculty
of Pharmacy, The University of Lahore, Lahore 54000, Pakistan
| |
Collapse
|
12
|
Wang B, Li D, Fiselier A, Kovalchuk I, Kovalchuk O. High-CBD cannabis extracts inhibit the expression of proinflammatory factors via miRNA-mediated silencing in human small intestinal epithelial cells. Heliyon 2023; 9:e18817. [PMID: 37664748 PMCID: PMC10468390 DOI: 10.1016/j.heliyon.2023.e18817] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 09/05/2023] Open
Abstract
The incidence of chronic inflammatory disorders and autoimmune diseases is rapidly growing. To date, the COVID-19 pandemic caused by SARS-CoV-2 has killed over 6,209,000 people globally, while no drug has been proven effective for the disease. Screening natural anti-inflammatory compounds for clinical application has drawn much attention. In this study, we showed that high-CBD cannabis extracts #1, #5, #7, #169, and #317 suppressed the levels of expression of proinflammatory cyclooxygenase 2 (COX2) and increased the expression of the anti-inflammatory suppressor of cytokine signaling 3 (SOCS3) in human small intestinal epithelial cells (HSIEC) in TNFα/IFNγ-triggered inflammation. We revealed that these extracts, with the exception of extract #169, also profoundly attenuated induction of proinflammatory cytokines interleukin-6 (IL-6) and/or IL-8 proteins through miR-760- and miR-302c-3p-mediated silencing. The prevalent components in extracts #1 and #7 influenced the levels of IL-8 both individually as well as in combination with each other. However, the high-dose cannabis extracts displayed an inhibitory effect in the growth of HSIEC cells. These results show that our high-CBD cannabis extracts decrease the levels of proinflammatory molecules COX2, IL-6, and IL-8 via transcriptional suppression or miRNA-mediated silencing, highlighting their potential against COVID-19-associated cytokine storm syndrome.
Collapse
Affiliation(s)
- Bo Wang
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Alberta, T1K 3M4, Canada
- Pathway Rx Inc., Calgary, Alberta, T3H 4Z2, Canada
| | - Dongping Li
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Alberta, T1K 3M4, Canada
- Pathway Rx Inc., Calgary, Alberta, T3H 4Z2, Canada
| | - Anna Fiselier
- Pathway Rx Inc., Calgary, Alberta, T3H 4Z2, Canada
- Swysh Inc., Calgary, Alberta, T3H 4Z2, Canada
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Alberta, T1K 3M4, Canada
- Pathway Rx Inc., Calgary, Alberta, T3H 4Z2, Canada
- Swysh Inc., Calgary, Alberta, T3H 4Z2, Canada
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Alberta, T1K 3M4, Canada
- Pathway Rx Inc., Calgary, Alberta, T3H 4Z2, Canada
- Swysh Inc., Calgary, Alberta, T3H 4Z2, Canada
| |
Collapse
|
13
|
Zhao Q, He L, Zhang J, Li H, Li W, Zhou Z, Li Y. MicroRNA-598 inhibition ameliorates LPS-induced acute lung injury in mice through upregulating Ebf1 expression. Histochem Cell Biol 2023:10.1007/s00418-023-02192-7. [PMID: 37115319 PMCID: PMC10141928 DOI: 10.1007/s00418-023-02192-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 04/29/2023]
Abstract
Acute lung injury is a critical acute respiratory distress syndrome (ARDS) with high morbidity and mortality. MicroRNAs (miRNAs) have been demonstrated to play important roles regulating acute lung injury development. In this study, we found that the expression of miR-598 was significantly upregulated in the lung tissues of mice with lipopolysaccharide (LPS)-induced acute lung injury. Both loss-of-function and gain-of-function studies were performed to evaluate the function of miR-598 in acute lung injury. The results showed that inhibition of miR-598 attenuated inflammatory response, oxidative stress, and lung injury in mice treated with LPS, while overexpression of miR-598 exacerbated the LPS-induced acute lung injury. Mechanistically, transcription factor Early B-cell Factor-1 (Ebf1) was predicted and validated as a downstream target of miR-598. Overexpression of Ebf1 attenuated LPS-induced production of inflammatory cytokine TNF-α and IL-6, ameliorated LPS-induced oxidative stress, promoted proliferation, and inhibited apoptosis in murine lung epithelial-15 (MLE-15) cells. Moreover, we demonstrated that Ebf1 knockdown abolished the protective effect of miR-598 inhibition in LPS-treated MLE-15 cells. In summary, miR-598 inhibition ameliorates LPS-induced acute lung injury in mice through upregulating Ebf1 expression, which might provide potential therapeutic treatment for acute lung injury.
Collapse
Affiliation(s)
- Qi Zhao
- XianYang Vocational Technical College, Tongyi Avenue, Fengxi New Town, Xixian, Xi'an, 712000, Shaanxi, China
| | - Lei He
- Pharmaceutical Factory of Shaanxi, University of Chinese Medicine, No. 1 Weiyang Middle Road, Qindu, Distrtict, Xianyang, Shaanxi, China
| | - Junwu Zhang
- Shaanxi University of Chinese Medicine, Xixian Avenue, Xi'an, 712046, Shaanxi, China.
| | - Hong Li
- Shaanxi University of Chinese Medicine, Xixian Avenue, Xi'an, 712046, Shaanxi, China.
| | - Wanying Li
- Pharmaceutical Factory of Shaanxi, University of Chinese Medicine, No. 1 Weiyang Middle Road, Qindu, Distrtict, Xianyang, Shaanxi, China
| | - Zhihui Zhou
- Second Affiliated Hospital of Shaanxi University of Chinese Medicine, No. 831, Longtaiguan Road, Fengxi New Town, Xixian, Xi'an, 712000, Shaanxi, China
| | - Yuanyuan Li
- Second Affiliated Hospital of Shaanxi University of Chinese Medicine, No. 831, Longtaiguan Road, Fengxi New Town, Xixian, Xi'an, 712000, Shaanxi, China
| |
Collapse
|
14
|
Ben Ammar R. Potential Effects of Geraniol on Cancer and Inflammation-Related Diseases: A Review of the Recent Research Findings. Molecules 2023; 28:molecules28093669. [PMID: 37175079 PMCID: PMC10180430 DOI: 10.3390/molecules28093669] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/01/2023] [Accepted: 04/17/2023] [Indexed: 05/15/2023] Open
Abstract
Geraniol (GNL), a natural monoterpene, is found in many essential oils of fruits, vegetables, and herbs, including lavender, citronella, lemongrass, and other medicinal and aromatic plants. GNL is commonly used by the cosmetic and food industries and has shown a wide spectrum of pharmacological activities including anti-inflammatory, anticancer, antimicrobial, antioxidant, and neuroprotective activities. It represents a potential anti-inflammatory agent and a promising cancer chemopreventive agent, as it has been found to be effective against a broad range of cancers, including colon, prostate, breast, lung, skin, kidney, liver, and pancreatic cancer. Moreover, GNL scavenges free radicals and preserves the activity of antioxidant enzymes. In addition, GNL induces apoptosis and cell cycle arrest, modulates multiple molecular targets, including p53 and STAT3, activates caspases, and modulates inflammation via transcriptional regulation. In the present study, different modes of action are described for GNL's activity against cancer and inflammatory diseases. This compound protects various antioxidant enzymes, such as catalase, glutathione-S-transferase, and glutathione peroxidase. Experiments using allergic encephalomyelitis, diabetes, asthma, and carcinogenesis models showed that GNL treatment had beneficial effects with low toxicity. GNL has been shown to be effective in animal models and tumor cell lines, but there have not been any clinical studies carried out for it. The aim of the present review is to provide updated data on the potential effects of GNL on cancer and inflammation, and to enhance our understanding of molecular targets, involved pathways, and the possible use of GNL for clinical studies and therapeutic purposes in the treatment of cancer and inflammation-related diseases.
Collapse
Affiliation(s)
- Rebai Ben Ammar
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Laboratory of Aromatic and Medicinal Plants, Center of Biotechnology of Borj-Cedria, Technopole of Borj-Cedria, P.O. Box 901, Hammam-Lif 2050, Tunisia
| |
Collapse
|
15
|
Li R, Ren T, Zeng J, Xu H. ALCAM Deficiency Alleviates LPS-Induced Acute Lung Injury by Inhibiting Inflammatory Response. Inflammation 2023; 46:688-699. [PMID: 36418761 DOI: 10.1007/s10753-022-01765-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022]
Abstract
We investigated the effects and underlying mechanisms of activated leukocyte adhesion molecule (ALCAM) on acute lung injury (ALI) by using lipopolysaccharide (LPS)-induced ALI animal model and LPS-induced inflammation in vitro. In LPS-stimulated mice, ALCAM deficiency relieved lung injury, which manifested as reduced pathological changes in the lung tissue, reduced pulmonary edema, and reduced vascular permeability. Furthermore, we demonstrated that ALCAM deficiency reduced the infiltration of inflammatory cells, including neutrophil, eosinophil, and macrophages; the release of inflammatory cytokines, including IL-1β, IL-6, TNF-α, and COX2; and reduced the protein level of TLR4/NF-κB pathway (TLR4, MyD88, p-IkBɑ, and p-NF-κB p65). We also demonstrated that ALCAM deficiency reduced the expression of oxidative stress-related proteins (Nrf-2, HO-1, and NQO-1) and endoplasmic reticulum stress-related proteins (CHOP, GRP78, ATF-6, and p-eIF2ɑ). In addition, in LPS-induced inflammation in vitro, ALCAM overexpression promoted inflammatory response, oxidative stress, and ER stress. We established that ALCAM deficiency can suppress the ALI process by reducing inflammatory response, oxidative stress, and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Ruirui Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Shihezi University School of Medicine, No. 107, Shibei 2Nd Road, Shihezi, 832008, Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Tao Ren
- Three Departments of Cardiology, The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, 832008, Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Jianqiong Zeng
- Cardiovascular Surgery CCU, Foshan First People's Hospital, Foshan, 528000, Guangdong, People's Republic of China
| | - Hang Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Shihezi University School of Medicine, No. 107, Shibei 2Nd Road, Shihezi, 832008, Xinjiang Uygur Autonomous Region, People's Republic of China.
| |
Collapse
|
16
|
Ma J, Xu Y, Zhang M, Li Y. Geraniol ameliorates acute liver failure induced by lipopolysaccharide/D-galactosamine via regulating macrophage polarization and NLRP3 inflammasome activation by PPAR-γ methylation Geraniol alleviates acute liver failure. Biochem Pharmacol 2023; 210:115467. [PMID: 36849063 DOI: 10.1016/j.bcp.2023.115467] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 03/01/2023]
Abstract
Geraniol (Ger), a natural acyclic monoterpene alcohol, has been reported to exert protective effects through anti-inflammation in Acute liver failure (ALF). However, its specific roles and precise mechanisms underlying anti-inflammatory effects in ALF have not yet fully explored. We aimed to investigated the hepatoprotective effects and mechanisms of Ger against ALF induced by lipopolysaccharide (LPS)/D-galactosamine (GaIN). In this study, the liver tissue and serum of LPS/D-GaIN-induced mice were collected. The degree of liver tissue injury was evaluated by HE and TUNEL staining. Serum levels of liver injury markers (ALT and AST) and inflammatory factors were measured by ELISA assays. PCR and western blotting were conducted to determine the expression of inflammatory cytokines, NLRP3 inflammasome-related proteins, PPAR-γ pathway-related proteins, DNA Methyltransferases and M1/M2 polarization cytokines. Immunofluorescence staining was used to assess the localization and expression of macrophage markers (F4/80 and CD86), NLRP3 and PPAR-γ. In vitro experiments were performed in macrophages stimulated with LPS with or without IFN-γ. Purification of macrophages and cell apoptosis was analyzed using flow cytometry. We found that Ger effectively alleviated ALF in mice, specified by the attenuation of liver tissue pathological damage, inhibition of ALT, AST and inflammatory factor levels, and inactivation of NLRP3 inflammasome. Meanwhile, downregulation M1 macrophage polarization may involve in the protective effects of Ger. In vitro, Ger reduced the activation of NLRP3 inflammasome and apoptosis through regulating PPAR-γ methylation by inhibiting M1 macrophage polarization. In conclusion, Ger protects against ALF through suppressing NLRP3 inflammasome-mediated inflammation and LPS-induced macrophage M1 polarization via modulating PPAR-γ methylation.
Collapse
Affiliation(s)
- Jing Ma
- Infectious Disease Department, The Second XIANGYA Hospital of Central South University, Changsha, Hunan, China
| | - Yun Xu
- Infectious Disease Department, The Second XIANGYA Hospital of Central South University, Changsha, Hunan, China
| | - Min Zhang
- Infectious Disease Department, The Second XIANGYA Hospital of Central South University, Changsha, Hunan, China
| | - Yi Li
- Infectious Disease Department, The Second XIANGYA Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
17
|
Haute GV, Luft C, Pedrazza L, Antunes GL, Silveira J, de Souza Basso B, Levorse VGS, Bastos MS, Melo D, Rodrigues KF, Garcia MC, da Costa MS, Matzenbacher LS, Kaiber DB, Donadio MVF, Gracia-Sancho J, de Oliveira JR. Simvastatin attenuates inflammatory process on LPS-induced acute lung injury in mice. Respir Physiol Neurobiol 2023; 309:104002. [PMID: 36566004 DOI: 10.1016/j.resp.2022.104002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/05/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Acute lung injury (ALI) is a disease of high prevalence and is characterized by the excessive production of inflammatory mediators in the lungs of people sick. Inflammation is the major characteristic of ALI and studies report that inhibition of inflammatory cytokines could be an alternative treatment. Statins such as Simvastatin (SV) are known to their use for cholesterol reduction but also for inflammatory and immunoregulatory processes. In this study, we evaluated the effects of SV on LPS-induced alveolar macrophages and in ALI mice model. Our study has demonstrated the protective effects of SV on LPS-activated alveolar macrophages RAW 264.7 and LPS-induced ALI in mice. SV treatment significantly inhibited the alveolar macrophages activation by decreasing the iNOS, IL-1β, and IL-6 gene expression in vitro and in vivo. The treatment also decreased the inflammatory cells migration and the cytokines gene expression. Our findings suggest that SV can act as an anti-inflammatory agent for acute lung injury.
Collapse
Affiliation(s)
- Gabriela Viegas Haute
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Carolina Luft
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Department of Psychology, Brock University, St. Catharines, Canada
| | - Leonardo Pedrazza
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Géssica Luana Antunes
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Josiane Silveira
- Neuroprotection and Neurometabolic Diseases Laboratory (Wyse's Lab), Department of Biochemistry, ICBS, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Bruno de Souza Basso
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Vitor Giancarlo Schneider Levorse
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Matheus Scherer Bastos
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Denizar Melo
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Ketlin Fernanda Rodrigues
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Maria Claudia Garcia
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Mariana Severo da Costa
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Lucas Strassburger Matzenbacher
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Daniela Benvenutti Kaiber
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Márcio Vinícius Fagundes Donadio
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Department of Physiotherapy, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Jordi Gracia-Sancho
- Barcelona Hepatic Hemodynamic Laboratory, IDIBAPS - Hospital Clinic de Barcelona - CIBEREHD, Barcelona, Spain; Hepatology, Department of Clinical Research, University of Bern, Switzerland
| | - Jarbas Rodrigues de Oliveira
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.
| |
Collapse
|
18
|
El-Shiekh RA, Nabil G, Shokry AA, Ahmed YH, Al-Hawshabi OSS, Abdel-Sattar E. Arabincoside B isolated from Caralluma arabica as a potential anti-pneumonitis in LPS mice model. Inflammopharmacology 2023; 31:1437-1447. [PMID: 36820943 PMCID: PMC9948789 DOI: 10.1007/s10787-023-01159-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023]
Abstract
Acute lung injury (ALI) is a life-threatening condition usually associated with poor therapeutic outcomes and a high mortality rate. Since 2019, the situation has worsened due to the COVID-19 pandemic. ALI had approximately 40% of deaths before COVID-19, mainly due to the dysfunction of the blood-gas barrier that led to lung edema, failure of gas exchange, and dyspnea. Many strategies have been taken to mitigate the disease condition, such as diuretics, surfactants, antioxidants, glucocorticoids, heparin, and ventilators with concomitant sedatives. However, until now, there is no available effective therapy for ALI. Thus, we are presenting a new compound termed Arabincoside B (AR-B), recently isolated from Caralluma arabica, to be tested in such conditions. For that, the lipopolysaccharide (LPS) mice model was used to investigate the capability of the AR-B compound to control the ALI compared to standard dexamethasone. The results showed that AR-B had a significant effect on retrieving ALI. A further mechanistic study carried out in the serum, lung homogenate, histological, and immunohistochemistry sections revealed that the AR-B either in 50 mg/kg or 75 mg/kg dose inhibited pro-inflammatory cytokines such as IL-6, IL-13, NF-κB, TNFα, and NO and stimulated regulatory cytokines IL-10. Moreover, AR-B showed a considerable potential to protect the pulmonary tissue against oxidative stress by decreasing MDA and increasing catalase and Nrf2. Also, the AR-B exhibited an anti-apoptotic effect on the lung epithelium, confirmed by reducing COX and BAX expression and upregulating Bcl-2 expression. These results pave its clinical application for ALI.
Collapse
Affiliation(s)
- Riham A. El-Shiekh
- grid.7776.10000 0004 0639 9286Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562 Egypt
| | - Ghazal Nabil
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| | - Aya A. Shokry
- grid.7776.10000 0004 0639 9286Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211 Egypt
| | - Yasmine H. Ahmed
- grid.7776.10000 0004 0639 9286Department of Cytology & Histology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211 Egypt
| | - Othman S. S. Al-Hawshabi
- grid.411125.20000 0001 2181 7851Department of Biology, Faculty of Science, University of Aden, Aden, Yemen
| | - Essam Abdel-Sattar
- grid.7776.10000 0004 0639 9286Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562 Egypt
| |
Collapse
|
19
|
Che HY, Zhou CH, Lyu CC, Meng Y, He YT, Wang HQ, Wu HY, Zhang JB, Yuan B. Allicin Alleviated LPS-Induced Mastitis via the TLR4/NF-κB Signaling Pathway in Bovine Mammary Epithelial Cells. Int J Mol Sci 2023; 24:ijms24043805. [PMID: 36835218 PMCID: PMC9962488 DOI: 10.3390/ijms24043805] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/10/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023] Open
Abstract
Dairy farming is the most important economic activity in animal husbandry. Mastitis is the most common disease in dairy cattle and has a significant impact on milk quality and yield. The natural extract allicin, which is the main active ingredient of the sulfur-containing organic compounds in garlic, has anti-inflammatory, anticancer, antioxidant, and antibacterial properties; however, the specific mechanism underlying its effect on mastitis in dairy cows needs to be determined. Therefore, in this study, whether allicin can reduce lipopolysaccharide (LPS)-induced inflammation in the mammary epithelium of dairy cows was investigated. A cellular model of mammary inflammation was established by pretreating bovine mammary epithelial cells (MAC-T) with 10 µg/mL LPS, and the cultures were then treated with varying concentrations of allicin (0, 1, 2.5, 5, and 7.5 µM) added to the culture medium. MAC-T cells were examined using RT-qPCR and Western blotting to determine the effect of allicin. Subsequently, the level of phosphorylated nuclear factor kappa-B (NF-κB) was measured to further explore the mechanism underlying the effect of allicin on bovine mammary epithelial cell inflammation. Treatment with 2.5 µM allicin considerably decreased the LPS-induced increase in the levels of the inflammatory cytokines interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-8 (IL-8), and tumor necrosis factor-α (TNF-α) and inhibited activation of the NOD-like receptor protein 3 (NLRP3) inflammasome in cow mammary epithelial cells. Further research revealed that allicin also inhibited the phosphorylation of inhibitors of nuclear factor kappa-B-α (IκB-α) and NF-κB p65. In mice, LPS-induced mastitis was also ameliorated by allicin. Therefore, we hypothesize that allicin alleviated LPS-induced inflammation in the mammary epithelial cells of cows probably by affecting the TLR4/NF-κB signaling pathway. Allicin will likely become an alternative to antibiotics for the treatment of mastitis in cows.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jia-Bao Zhang
- Correspondence: (J.-B.Z.); (B.Y.); Tel.: +86-431-8783-6551 (J.-B.Z.); +86-431-8783-6536 (B.Y.)
| | - Bao Yuan
- Correspondence: (J.-B.Z.); (B.Y.); Tel.: +86-431-8783-6551 (J.-B.Z.); +86-431-8783-6536 (B.Y.)
| |
Collapse
|
20
|
Correa N, Orlando R. Extract of Laurus nobilis attenuates inflammation and epithelial ulcerations in an experimental model of inflammatory bowel disease. WIKIJOURNAL OF MEDICINE 2023. [DOI: 10.15347/wjm/2023.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Inflammatory bowel diseases (IBD), including Crohn's disease and ulcerative colitis, are classified as chronic inflammatory disorders and typically require anti-inflammatory drug therapies, such as glucocorticoid regimens, non-steroidal anti-inflammatory drugs, and biologics, aimed at reducing inflammation in the bowel wall. However, each of these therapies is accompanied by a list of possible serious side effects. Because of this, there remains an urgent need to identify new pharmacologic options to reduce or prevent the pro-inflammatory events of IBD while minimizing adverse side effects, and to make available more cost-effective treatment modalities. We have previously identified several herbal extracts that demonstrate potent bio-inhibitory activity of the innate immune response. In particular, Laurus nobilis (LN), or more commonly called bay laurel, demonstrated significant anti-inflammatory function by inhibiting nuclear factor-κB activation. Based upon our original in vitro findings, we have now examined the effects of this herbal extract on a murine dextran sodium sulfate (DSS) model of IBD. Hematoxylin and eosin-stained paraffin sections prepared from DSS treated animals show clear epithelial damage, including ulcerations, extensive neutrophil infiltration into the mucosal layer, and granuloma formation. Tissue from DSS treated animals that also received LN extract showed improved tissue morphology more closely resembling that from control animals. In addition, DSS treated mice with co-administration of LN extract showed a significant reduction in CD4+ antibody staining within the mucosal layer in colonic sections indicating reduced lymphocyte infiltration. Based on these findings, we believe that administration of LN extracts may be effective in reducing the intestinal epithelial damage seen in human IBD and warrants further investigation through clinical trials. Lay Summary: Inflammatory bowel diseases (IBD), such as Crohn's disease (CD) and ulcerative colitis (UC), manifest as chronic inflammation and ulceration of tissues lining the digestive tract. CD involves inflammation of the deeper layers of the digestive tract, including both the small and large intestines, and less commonly, the upper digestive tract. UC involves inflammation along the lining of the colon and rectum. Steroid or biologic treatments for IBD are common, however, are limited due to significant side effects and/or prohibitive cost. In the present study, we provide evidence for use of the natural product, Laurus nobilis (bay leaf), as a safe and effective anti-inflammatory therapy for IBD.
Collapse
|
21
|
Guo B, Zuo Z, Di X, Huang Y, Gong G, Xu B, Wang L, Zhang X, Liang Z, Hou Y, Liu X, Hu Z. Salidroside attenuates HALI via IL-17A-mediated ferroptosis of alveolar epithelial cells by regulating Act1-TRAF6-p38 MAPK pathway. Cell Commun Signal 2022; 20:183. [PMID: 36411467 PMCID: PMC9677645 DOI: 10.1186/s12964-022-00994-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 10/24/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Hyperoxia-induced acute lung injury (HALI) is a critical life-threatening disorder characterized by severe infiltration immune cells and death of type II alveolar epithelial cells (AECII). However, little is known about the relations between immune cells and AECII in HALI. IL-17A is a pro-inflammatory cytokine mainly secreted by Th17 cells, contributing to the pathogenesis of various inflammatory diseases. The present study investigated the role of IL-17A in cell-cell communication between immune cells and AECII in HALI, and explored the therapeutic effect of salidroside (Sal, a natural anti-inflammatory agents) on HALI. METHODS Mice with HALI were induced by exposure to hyperoxia over 90% for 12 h, 24 h, 48 h or 72 h, and the optimal timing was detected by H&E and Masson staining. Ferroptosis was confirmed by detecting the levels of MDA, Fe2+ and GPX4, and the morphological alterations of AECII under transmission electron microscopy. The expression of pro-inflammatory cytokine, including IL-6, TGF-β1, IL-17A and IL-17A receptor (IL-17RA) were measured by Western blotting and immunohistochemical stanning. The ferroptosis-related Act1/TRAF6/p38 MAPK pathway was detected by Western blotting. The role of pro-inflammatory cytokine IL-17A for AECII ferroptosis, and the effect of Sal on HALI were investigated by administration of Y-320 (IL-17 inhibitor) and Sal respectively 3 days before mice exposed to hyperoxia. RESULTS Mice exposed to hyperoxia for 24 h suffered sufficient HALI with inflammatory cell infiltration and collagen deposition, and exhibited features of ferroptosis under TME. Meanwhile, compared with sham mice, mice exposed to hyperoxia showed down-regulation of GPX4, and up-regulation of IL-6, TGF-β1, IL-17A, IL-17RA, Act1, TRAF6, p38 MAPK and p-p38 MAPK. Moreover, inhibition of IL-17A with Y-320 or administration with Sal could reverse the effect caused by hyperoxia respectively. CONCLUSIONS IL-17A is associated with immune cells infiltration in HALI, and contributes to ferroptosis of AECII that related to Act1/TRAF6/p38 MAPK pathway. Additionally, Sal protects against HALI throughout the whole pathogenic process. Video Abstract.
Collapse
Affiliation(s)
- Baoyue Guo
- grid.454145.50000 0000 9860 0426The Intensive Care Unit, The First Affiliated Hospital, Jinzhou Medical University, No. 2, Section 5, Renmin Street, Jinzhou, 121012 People’s Republic of China
| | - Zhongfu Zuo
- grid.454145.50000 0000 9860 0426Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001 People’s Republic of China
| | - Xingwei Di
- grid.454145.50000 0000 9860 0426The Intensive Care Unit, The First Affiliated Hospital, Jinzhou Medical University, No. 2, Section 5, Renmin Street, Jinzhou, 121012 People’s Republic of China
| | - Ying Huang
- grid.454145.50000 0000 9860 0426Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001 People’s Republic of China
| | - Gu Gong
- The Intensive Care Unit, The Central Hospital of Jinzhou, Jinzhou, 121001 People’s Republic of China
| | - Bo Xu
- grid.454145.50000 0000 9860 0426The Intensive Care Unit, The First Affiliated Hospital, Jinzhou Medical University, No. 2, Section 5, Renmin Street, Jinzhou, 121012 People’s Republic of China
| | - Lulu Wang
- The Intensive Care Unit, The Central Hospital of Jinzhou, Jinzhou, 121001 People’s Republic of China
| | - Xiaoyu Zhang
- The Intensive Care Unit, The Central Hospital of Dandong, Dandong, 118002 People’s Republic of China
| | - Zhuang Liang
- grid.454145.50000 0000 9860 0426The Intensive Care Unit, The First Affiliated Hospital, Jinzhou Medical University, No. 2, Section 5, Renmin Street, Jinzhou, 121012 People’s Republic of China
| | - Yang Hou
- grid.454145.50000 0000 9860 0426College of Biological Information Engineering, Jinzhou Medical University, Jinzhou, 121001 People’s Republic of China
| | - Xuezheng Liu
- grid.454145.50000 0000 9860 0426Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001 People’s Republic of China
| | - Zhansheng Hu
- grid.454145.50000 0000 9860 0426The Intensive Care Unit, The First Affiliated Hospital, Jinzhou Medical University, No. 2, Section 5, Renmin Street, Jinzhou, 121012 People’s Republic of China
| |
Collapse
|
22
|
Yu H, Ju Q, Cheng S. Regulating Function of miR-146a Derived from Bone Marrow Mesenchymal Stem Cell (BMSC) in Acute Lung Injury. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
This study assesses the mechanism of miR-146a derived from Bone marrow mesenchymal stem cell (BMSC) in acute lung injury. The model of ALI rats was established through endotracheal perfusion of LPS followed by analysis histological changes by HE staining. The source of BMSC was detected
through flow cytometry and change of miRNA was detected through Array method. The miR-146a level in lung tissue was detected with RT-PCR and expression of Bcl-2, Bax and Capase-9 was detected with IF and Western Blot. A high expression of CD90 and CD105 was found in BMSC with negative CD11bc
and CD34 level. 39 downregulated miRNAs and 20 upregulated miRNAs were found in ALI with miR-146a being the most significant. The apoptotic level induced with LPS could be restrained by miR-146a. In addition, miR-146a could upregulate Bcl-2 and downregulate Bax and Caspase-9. In conclusion,
ALI could be restrained by the low expression of miR-146a.
Collapse
Affiliation(s)
- Honglei Yu
- Department of Pediatrics, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei, 437000, China
| | - Qiu Ju
- Department of Pediatrics, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei, 437000, China
| | - Shouchao Cheng
- Department of Pediatrics, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei, 437000, China
| |
Collapse
|
23
|
Analysis of Anti-Cancer and Anti-Inflammatory Properties of 25 High-THC Cannabis Extracts. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27186057. [PMID: 36144796 PMCID: PMC9506243 DOI: 10.3390/molecules27186057] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 12/24/2022]
Abstract
Cannabis sativa is one of the oldest cultivated plants. Many of the medicinal properties of cannabis are known, although very few cannabis-based formulations became prescribed drugs. Previous research demonstrated that cannabis varieties are very different in their medicinal properties, likely due to the entourage effect-the synergistic or antagonistic effect of various cannabinoids and terpenes. In this work, we analyzed 25 cannabis extracts containing high levels of delta-9-tetrahydrocannabinol (THC). We used HCC1806 squamous cell carcinoma and demonstrated various degrees of efficiency of the tested extracts, from 66% to 92% of growth inhibition of cancer cells. Inflammation was tested by induction of inflammation with TNF-α/IFN-γ in WI38 human lung fibroblasts. The efficiency of the extracts was tested by analyzing the expression of COX2 and IL6; while some extracts aggravated inflammation by increasing the expression of COX2/IL6 by 2-fold, other extracts decreased inflammation, reducing expression of cytokines by over 5-fold. We next analyzed the level of THC, CBD, CBG and CBN and twenty major terpenes and performed clustering and association analysis between the chemical composition of the extracts and their efficiency in inhibiting cancer growth and curbing inflammation. A positive correlation was found between the presence of terpinene (pval = 0.002) and anti-cancer property; eucalyptol came second, with pval of 0.094. p-cymene and β-myrcene positively correlated with the inhibition of IL6 expression, while camphor correlated negatively. No significant correlation was found for COX2. We then performed a correlation analysis between cannabinoids and terpenes and found a positive correlation for the following pairs: α-pinene vs. CBD, p-cymene vs. CBGA, terpenolene vs. CBGA and isopulegol vs. CBGA. Our work, thus, showed that most of high-THC extracts demonstrate anti-cancer activity, while only certain selected extracts showed anti-inflammatory activity. Presence of certain terpenes, such as terpinene, eucalyptol, cymene, myrcene and camphor, appear to have modulating effects on the activity of cannabinoids.
Collapse
|
24
|
Renal Ischemia/Reperfusion Mitigation via Geraniol: The Role of Nrf-2/HO-1/NQO-1 and TLR2,4/MYD88/NFκB Pathway. Antioxidants (Basel) 2022; 11:antiox11081568. [PMID: 36009287 PMCID: PMC9405463 DOI: 10.3390/antiox11081568] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/30/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Renal ischemia/reperfusion injury is a clinically recurrent event during kidney transplantation. Geraniol is a natural monoterpene essential oil component. This study aimed to inspect geraniol's reno-protective actions against renal I/R injury with further analysis of embedded mechanisms of action through scrutinizing the Nrf-2/HO-1/NQO-1 and TLR2,4/MYD88/NFκB signaling pathways. METHODS Wistar male rats were randomized into five groups: Sham, Sham + geraniol, Renal I/R, and two Renal I/R + geraniol groups representing two doses of geraniol (100 and 200 mg/kg) for 14 days before the renal I/R. Renal I/R was surgically induced by occluding both left and right renal pedicles for 45 min, followed by reperfusion for 24 h. A docking study was performed to anticipate the expected affinity of geraniol towards three protein targets: hTLR4/MD2, hTLR2, and hNrf2/Keap1. RESULTS Renal I/R rats experienced severely compromised renal functions, histological alteration, oxidative stress status, escalated Nrf-2/HO-1/NQO-1, and amplified TLR2,4/MYD88/NFκB. Geraniol administration ameliorated renal function, alleviated histological changes, and enhanced Nrf-2/HO-1/NQO-1 with a subsequent intensification of antioxidant enzyme activities. Geraniol declined TLR2,4/MYD88/NFκB with subsequent TNF-α, IFN-γ, MCP-1 drop, Bax, caspase-3, and caspase-9 reduction IL-10 and Bcl-2 augmentation. Geraniol exhibited good fitting in the binding sites of the three in silico examined targets. CONCLUSIONS Geraniol might protect against renal I/R via the inhibition of the TLR2,4/MYD88/NFκB pathway, mediating anti-inflammation and activation of the Nrf2 pathway, intervening in antioxidative activities.
Collapse
|
25
|
Theivendran S, Gu Z, Tang J, Yang Y, Song H, Yang Y, Zhang M, Cheng D, Yu C. Nanostructured Organosilica Nitric Oxide Donors Intrinsically Regulate Macrophage Polarization with Antitumor Effect. ACS NANO 2022; 16:10943-10957. [PMID: 35735363 DOI: 10.1021/acsnano.2c03348] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Nitric oxide (NO) has many important biological functions; however, it has been a long-standing challenge to utilize the exogenous NO donor itself in the activation of macrophages for cancer immunotherapy. Herein, we report the synthesis of a nanoparticle-based NO delivery platform with a rational design for effective NO delivery and macrophage activation. S-Nitrosothiol (SNO) modified organosilica nanoparticles with a tetrasulfide-containing composition produced a higher level of intracellular NO than their bare silica counterparts in macrophages. Enhanced intracellular delivery of NO resulted in mitochondrial dysfunction and disruption of the tricarboxylic acid cycle, leading to macrophage activation and delayed tumor growth. This study provides insights on intracellularly delivered NO for regulating the polarization of macrophages and cancer immunotherapy.
Collapse
Affiliation(s)
- Shevanuja Theivendran
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Zhengying Gu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Jie Tang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yannan Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yang Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Min Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Dan Cheng
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| |
Collapse
|
26
|
Havermans A, Mallock N, Zervas E, Caillé-Garnier S, Mansuy T, Michel C, Pennings JLA, Schulz T, Schwarze PE, Solimini R, Tassin JP, Vardavas CI, Merino M, Pauwels CGGM, van Nierop LE, Lambré C, Bolling AK. Review of industry reports on EU priority tobacco additives part A: Main outcomes and conclusions. Tob Prev Cessat 2022; 8:27. [PMID: 35860504 PMCID: PMC9255285 DOI: 10.18332/tpc/151529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 02/01/2023]
Abstract
The European Union Tobacco Products Directive (EU TPD) mandates enhanced reporting obligations for tobacco manufacturers regarding 15 priority additives. Within the Joint Action on Tobacco Control (JATC), a review panel of independent experts was appointed for the scientific evaluation of the additive reports submitted by a consortium of 12 tobacco manufacturers. As required by the TPD, the reports were evaluated based on their comprehensiveness, methodology and conclusions. In addition, we evaluated the chemical, toxicological, addictive, inhalation facilitating and flavoring properties of the priority additives based on the submitted reports, supplemented by the panel's expert knowledge and some independent literature. The industry concluded that none of the additives is associated with concern. Due to significant methodological limitations, we question the scientific validity of these conclusions and conclude that they are not warranted. Our review demonstrates that many issues regarding toxicity, addictiveness and attractiveness of the additives have not been sufficiently addressed, and therefore concerns remain. For example, menthol facilitates inhalation by activation of the cooling receptor TRPM8. The addition of sorbitol and guar gum leads to a significant increase of aldehydes that may contribute to toxicity and addictiveness. Titanium dioxide particles (aerodynamic diameter <10 µm) are legally classified as carcinogenic when inhaled. For diacetyl no report was provided. Overall, the industry reports were not comprehensive, and the information presented provides an insufficient basis for the regulation of most additives. We, therefore, advise MS to consider alternative approaches such as the precautionary principle.
Collapse
Affiliation(s)
- Anne Havermans
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Nadja Mallock
- German Federal Institute for Risk Assessment, Berlin, Germany
| | - Efthimios Zervas
- Hellenic Thoracic Society, Athens, Greece
- School of Applied Arts and Sustainable Design, Hellenic Open University, Athens, Greece
| | | | - Thibault Mansuy
- French Agency for Food, Environmental and Occupational Health and Safety, Paris, France
| | - Cécile Michel
- French Agency for Food, Environmental and Occupational Health and Safety, Paris, France
| | - Jeroen L. A. Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Thomas Schulz
- German Federal Institute for Risk Assessment, Berlin, Germany
| | | | | | | | | | - Miguel Merino
- Andalusia Agency For Agriculture and Fisheries Development, Seville, Spain
| | - Charlotte G. G. M. Pauwels
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Lotte E. van Nierop
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Claude Lambré
- National Institute of Health and Medical Research, Paris, France
| | | |
Collapse
|
27
|
Csikós E, Csekő K, Kemény Á, Draskóczi L, Kereskai L, Kocsis B, Böszörményi A, Helyes Z, Horváth G. Pinus sylvestris L. and Syzygium aromaticum (L.) Merr. & L. M. Perry Essential Oils Inhibit Endotoxin-Induced Airway Hyperreactivity despite Aggravated Inflammatory Mechanisms in Mice. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123868. [PMID: 35744988 PMCID: PMC9229653 DOI: 10.3390/molecules27123868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/10/2022] [Accepted: 06/11/2022] [Indexed: 11/16/2022]
Abstract
Scots pine (SO) and clove (CO) essential oils (EOs) are commonly used by inhalation, and their main components are shown to reduce inflammatory mediator production. The aim of our research was to investigate the chemical composition of commercially available SO and CO by gas chromatography–mass spectrometry and study their effects on airway functions and inflammation in an acute pneumonitis mouse model. Inflammation was evoked by intratracheal endotoxin and EOs were inhaled three times during the 24 h experimental period. Respiratory function was analyzed by unrestrained whole-body plethysmography, lung inflammation by semiquantitative histopathological scoring, myeloperoxidase (MPO) activity and cytokine measurements. α-Pinene (39.4%) was the main component in SO, and eugenol (88.6%) in CO. Both SO and CO significantly reduced airway hyperresponsiveness, and prevented peak expiratory flow, tidal volume increases and perivascular edema formation. Meanwhile, inflammatory cell infiltration was not remarkably affected. In contrast, MPO activity and several inflammatory cytokines (IL-1β, KC, MCP-1, MIP-2, TNF-α) were aggravated by both EOs. This is the first evidence that SO and CO inhalation improve airway function, but enhance certain inflammatory parameters. These results suggest that these EOs should be used with caution in cases of inflammation-associated respiratory diseases.
Collapse
Affiliation(s)
- Eszter Csikós
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pecs, H-7624 Pecs, Hungary;
| | - Kata Csekő
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pecs, H-7624 Pecs, Hungary; (K.C.); (Á.K.); (L.D.); (Z.H.)
- Szentágothai Research Centre, University of Pecs, H-7624 Pecs, Hungary
| | - Ágnes Kemény
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pecs, H-7624 Pecs, Hungary; (K.C.); (Á.K.); (L.D.); (Z.H.)
- Szentágothai Research Centre, University of Pecs, H-7624 Pecs, Hungary
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pecs, H-7624 Pecs, Hungary
| | - Lilla Draskóczi
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pecs, H-7624 Pecs, Hungary; (K.C.); (Á.K.); (L.D.); (Z.H.)
- Szentágothai Research Centre, University of Pecs, H-7624 Pecs, Hungary
| | - László Kereskai
- Department of Pathology, Medical School, University of Pecs, H-7624 Pecs, Hungary;
| | - Béla Kocsis
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, H-7624 Pecs, Hungary;
| | - Andrea Böszörményi
- Institute of Pharmacognosy, Faculty of Pharmacy, Semmelweis University, H-1085 Budapest, Hungary;
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pecs, H-7624 Pecs, Hungary; (K.C.); (Á.K.); (L.D.); (Z.H.)
- Szentágothai Research Centre, University of Pecs, H-7624 Pecs, Hungary
- PharmInVivo Ltd., H-7629 Pecs, Hungary
| | - Györgyi Horváth
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pecs, H-7624 Pecs, Hungary;
- Correspondence: ; Tel.: +36-72-503650-28823
| |
Collapse
|
28
|
Jang WY, Lee HP, Kim SA, Huang L, Yoon JH, Shin CY, Mitra A, Kim HG, Cho JY. Angiopteris cochinchinensis de Vriese Ameliorates LPS-Induced Acute Lung Injury via Src Inhibition. PLANTS 2022; 11:plants11101306. [PMID: 35631731 PMCID: PMC9143704 DOI: 10.3390/plants11101306] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022]
Abstract
Growing demand for treatment options against acute lung injury (ALI) emphasizes studies on plant extracts harboring anti-inflammatory effects. According to GC-MS analysis, Angiopteris cochinchinensis de Vriese consists of various flavonoids with anti-inflammatory activities. Thus, in this study, the anti-inflammatory effects of an extract of Angiopteris cochinchinensis de Vriese (Ac-EE) were assessed using RAW264.6 murine macrophages and a lipopolysaccharide (LPS)-induced ALI model. Ac-EE reduced the nitric oxide production in murine macrophages increased by LPS induction. Moreover, protective effects of Ac-EE on lung tissue were demonstrated by shrinkage of edema and lung injury. Reduced neutrophil infiltration and formation of hyaline membranes were also detected in lung tissues after H&E staining. Semiquantitative RT-PCR, quantitative real-time PCR, and ELISA showed that Ac-EE inhibits the production of proinflammatory mediators, including iNOS and COX-2, and cytokines, such as TNF-α, IL-1β, and IL-6. An Ac-EE-mediated anti-inflammatory response was derived from inhibiting the NF-κB signaling pathway, which was evaluated by luciferase reporter assay and Western blotting analysis. A cellular thermal shift assay revealed that the prime target of Ac-EE in alleviating inflammation was Src. With its direct binding with Src, Angiopteris cochinchinensis de Vriese significantly mitigates lung injury, showing possibilities of its potential as an effective botanical drug.
Collapse
Affiliation(s)
- Won Young Jang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (W.Y.J.); (H.P.L.); (S.A.K.)
| | - Hwa Pyoung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (W.Y.J.); (H.P.L.); (S.A.K.)
| | - Seung A Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (W.Y.J.); (H.P.L.); (S.A.K.)
| | - Lei Huang
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea; (L.H.); (J.H.Y.); (C.Y.S.)
| | - Ji Hye Yoon
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea; (L.H.); (J.H.Y.); (C.Y.S.)
| | - Chae Yun Shin
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea; (L.H.); (J.H.Y.); (C.Y.S.)
| | - Ankita Mitra
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea;
| | - Han Gyung Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (W.Y.J.); (H.P.L.); (S.A.K.)
- Correspondence: (H.G.K.); (J.Y.C.); Tel.: +82-31-290-7878 (H.G.K.); +82-31-290-7868 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (W.Y.J.); (H.P.L.); (S.A.K.)
- Correspondence: (H.G.K.); (J.Y.C.); Tel.: +82-31-290-7878 (H.G.K.); +82-31-290-7868 (J.Y.C.)
| |
Collapse
|
29
|
Lei CX, Xie YJ, Li SJ, Jiang P, Du JX, Tian JJ. Fabp4 contributes toward regulating inflammatory gene expression and oxidative stress in Ctenopharyngodon idella. Comp Biochem Physiol B Biochem Mol Biol 2022; 259:110715. [PMID: 34999220 DOI: 10.1016/j.cbpb.2022.110715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/20/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022]
Abstract
Fatty acid-binding protein (Fabp)-4 is a member of the FABP family. Mammalian fabp4 has been demonstrated to involve in inflammation and immunity, whereas the related data of fish fabp4 remain limited. Therefore, we further investigated the effects of fabp4 on immunity in Ctenopharyngodon idella. The fabp4 sequence spanned 405 bp was cloned first, sharing high identity to fabp4 from other fish and mammals. Fabp4 expression was the highest in the adipose tissue, followed by the heart, muscle, and liver. In vivo, lipopolysaccharide (LPS) triggered the expression of fabp4, toll-like receptor (tlr)-22, interleukin (il)-1β, and tumor necrosis factor (tnf)-α in the kidney and spleen. In vitro, exposing C. idella CIK cells to LPS decreased their viability, and the expression of fabp4 was also increased by LPS. However, BMS309403, an inhibitor of FABP4, mitigated these effects. Furthermore, treating the cells with LPS or fabp4 overexpression plasmids resulted in reactive oxygen species (ROS) generation and upregulation of inflammatory genes expression, including tlr22, type-I interferon (ifn-1), interferon regulatory factor (irf)-7, tnfα, il-1β, and interferon-β promoter stimulator 1. These effects were ameliorated by preincubation with BMS309403. Moreover, incubating the cells with glutathione reduced the production of ROS and the expression of inflammatory genes that were evoked by LPS and plasmid treatments. These results showed that fabp4 acts as a pro-inflammatory molecule via elevating ROS levels, providing a novel understanding of the molecular regulation of innate immunity in teleosts.
Collapse
Affiliation(s)
- Cai-Xia Lei
- Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, PR China; Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, PR China
| | - Yu-Jing Xie
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, PR China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, PR China
| | - Sheng-Jie Li
- Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, PR China; Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, PR China.
| | - Peng Jiang
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, PR China
| | - Jin-Xing Du
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, PR China
| | - Jing-Jing Tian
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, PR China
| |
Collapse
|
30
|
New AKT-dependent mechanisms of anti-COVID-19 action of high-CBD Cannabis sativa extracts. Cell Death Dis 2022; 8:110. [PMID: 35277472 PMCID: PMC8913855 DOI: 10.1038/s41420-022-00876-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 02/05/2022] [Accepted: 02/10/2022] [Indexed: 02/07/2023]
Abstract
COVID-19 is caused by the SARS-CoV-2 virus, which enters target cells via interactions with ACE2 and TMPRSS2. Here, we show AKT serine/threonine kinase-dependent epigenetic control of ACE2 and TMPRSS2 expression by high-cannabidiol (CBD) cannabis extracts and their individual components. CBD alone and extracts #1, #5, #7, and #129 downregulated ACE2 and TMPRSS2 in lung fibroblast WI-38 cells through AKT-mediated inhibition. miR-200c-3p and let-7a-5p were two contributing miRNAs in CBD-mediated suppression of ACE2 and TMPRSS2. CBD and terpene PTWT2.2 profoundly inhibited ACE2 and TMPRSS2 expression, both individually and in combination. Extracts #1, #5, #7, and #169 suppressed COX2 expression and remarkably attenuated TNFα/IFNγ-triggered induction of proinflammatory factors IL-6 and IL-8 by AKT pathway. The most abundant molecules present in extracts #1 and #7 modulated the expression of COX2, IL-6, and IL-8 both individually and in combination. These results reveal that high-CBD cannabis extracts attenuated ACE2 and TMPRSS2 expression and the induction of inflammatory mediators COX2, IL-6, and IL-8 via the AKT pathway, highlighting their potential anti-COVID-19 features.
Collapse
|
31
|
Amelioration of Endotoxin-Induced Acute Lung Injury and Alveolar Epithelial Cells Apoptosis by Simvastatin Is Associated with Up-Regulation of Survivin/NF-kB/p65 Pathway. Int J Mol Sci 2022; 23:ijms23052596. [PMID: 35269738 PMCID: PMC8910433 DOI: 10.3390/ijms23052596] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/02/2022] [Accepted: 02/06/2022] [Indexed: 02/05/2023] Open
Abstract
Disruption of the alveolar−endothelial barrier caused by inflammation leads to the progression of septic acute lung injury (ALI). In the present study, we investigated the beneficial effects of simvastatin on the endotoxin lipopolysaccharide (LPS)-induced ALI and its related mechanisms. A model of ALI was induced within experimental sepsis developed by intraperitoneal injection of a single non-lethal LPS dose after short-term simvastatin pretreatment (10−40 mg/kg orally). The severity of the lung tissue inflammatory injury was expressed as pulmonary damage scores (PDS). Alveolar epithelial cell apoptosis was confirmed by TUNEL assay (DNA fragmentation) and expressed as an apoptotic index (AI), and immunohistochemically for cleaved caspase-3, cytochrome C, and anti-apoptotic Bcl-xL, an inhibitor of apoptosis, survivin, and transcriptional factor, NF-kB/p65. Severe inflammatory injury of pulmonary parenchyma (PDS 3.33 ± 0.48) was developed after the LPS challenge, whereas simvastatin significantly and dose-dependently protected lung histology after LPS (p < 0.01). Simvastatin in a dose of 40 mg/kg showed the most significant effects in amelioration alveolar epithelial cells apoptosis, demonstrating this as a marked decrease of AI (p < 0.01 vs. LPS), cytochrome C, and cleaved caspase-3 expression. Furthermore, simvastatin significantly enhanced the expression of Bcl-xL and survivin. Finally, the expression of survivin and its regulator NF-kB/p65 in the alveolar epithelium was in strong positive correlation across the groups. Simvastatin could play a protective role against LPS-induced ALI and apoptosis of the alveolar−endothelial barrier. Taken together, these effects were seemingly mediated by inhibition of caspase 3 and cytochrome C, a finding that might be associated with the up-regulation of cell-survival survivin/NF-kB/p65 pathway and Bcl-xL.
Collapse
|
32
|
Geraniol Averts Methotrexate-Induced Acute Kidney Injury via Keap1/Nrf2/HO-1 and MAPK/NF-κB Pathways. Curr Issues Mol Biol 2021; 43:1741-1755. [PMID: 34889889 PMCID: PMC8929074 DOI: 10.3390/cimb43030123] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 01/05/2023] Open
Abstract
Objectives: Geraniol, a natural monoterpene, is an essential oil component of many plants. Methotrexate is an anti-metabolite drug, used for cancer and autoimmune conditions; however, clinical uses of methotrexate are limited by its concomitant renal injury. This study investigated the efficacy of geraniol to prevent methotrexate-induced acute kidney injury and via scrutinizing the Keap1/Nrf2/HO-1, P38MAPK/NF-κB and Bax/Bcl2/caspase-3 and -9 pathways. Methods: Male Wister rats were allocated into five groups: control, geraniol (orally), methotrexate (IP), methotrexate and geraniol (100 and 200 mg/kg). Results: Geraniol effectively reduced the serum levels of creatinine, urea and Kim-1 with an increase in the serum level of albumin when compared to the methotrexate-treated group. Geraniol reduced Keap1, escalated Nrf2 and HO-1, enhanced the antioxidant parameters GSH, SOD, CAT and GSHPx and reduced MDA and NO. Geraniol decreased renal P38 MAPK and NF-κB and ameliorated the inflammatory mediators TNF-α, IL-1β, IL-6 and IL-10. Geraniol negatively regulated the apoptotic mediators Bax and caspase-3 and -9 and increased Bcl2. All the biochemical findings were supported by the alleviation of histopathological changes in kidney tissues. Conclusion: The current findings support that co-administration of geraniol with methotrexate may attenuate methotrexate-induced acute kidney injury.
Collapse
|
33
|
Timalsina D, Pokhrel KP, Bhusal D. Pharmacologic Activities of Plant-Derived Natural Products on Respiratory Diseases and Inflammations. BIOMED RESEARCH INTERNATIONAL 2021; 2021:1636816. [PMID: 34646882 PMCID: PMC8505070 DOI: 10.1155/2021/1636816] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022]
Abstract
Respiratory inflammation is caused by an air-mediated disease induced by polluted air, smoke, bacteria, and viruses. The COVID-19 pandemic is also a kind of respiratory disease, induced by a virus causing a serious effect on the lungs, bronchioles, and pharynges that results in oxygen deficiency. Extensive research has been conducted to find out the potent natural products that help to prevent, treat, and manage respiratory diseases. Traditionally, wider floras were reported to be used, such as Morus alba, Artemisia indica, Azadirachta indica, Calotropis gigantea, but only some of the potent compounds from some of the plants have been scientifically validated. Plant-derived natural products such as colchicine, zingerone, forsythiaside A, mangiferin, glycyrrhizin, curcumin, and many other compounds are found to have a promising effect on treating and managing respiratory inflammation. In this review, current clinically approved drugs along with the efficacy and side effects have been studied. The study also focuses on the traditional uses of medicinal plants on reducing respiratory complications and their bioactive phytoconstituents. The pharmacological evidence of lowering respiratory complications by plant-derived natural products has been critically studied with detailed mechanism and action. However, the scientific validation of such compounds requires clinical study and evidence on animal and human models to replace modern commercial medicine.
Collapse
Affiliation(s)
- Deepak Timalsina
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal
| | | | - Deepti Bhusal
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal
| |
Collapse
|
34
|
Liu Y, Ma X, Liang H, Stephanopoulos G, Zhou K. Monoterpenoid biosynthesis by engineered microbes. J Ind Microbiol Biotechnol 2021; 48:6380491. [PMID: 34601590 DOI: 10.1093/jimb/kuab065] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 08/27/2021] [Indexed: 11/14/2022]
Abstract
Monoterpenoids are C10 isoprenoids and constitute a large family of natural products. They have been used as ingredients in food, cosmetics and therapeutic products. Many monoterpenoids such as linalool, geraniol, limonene and pinene are volatile and can be found in plant essential oils. Conventionally, these bioactive compounds are obtained from plant extracts by using organic solvents or by distillation method, which are costly and laborious if high purity product is desired. In recent years, microbial biosynthesis has emerged as alternative source of monoterpenoids with great promise for meeting the increasing global demand for these compounds. However, current methods of production are not yet at levels required for commercialization. Production efficiency of monoterpenoids in microbial hosts is often restricted by high volatility of the monoterpenoids, a lack of enzymatic activity and selectivity, and/or product cytotoxicity to the microbial hosts. In this review, we summarize advances in microbial production of monoterpenoids over the past three years with particular focus on the key metabolic engineering strategies for different monoterpenoid products. We also provide our perspective on the promise of future endeavors to improve monoterpenoid productivity.
Collapse
Affiliation(s)
- Yurou Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore.,Disruptive & Sustainable Technologies for Agricultural Precision, Singapore-MIT Alliance for Research and Technology, Singapore
| | - Xiaoqiang Ma
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore
| | - Hong Liang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore.,Disruptive & Sustainable Technologies for Agricultural Precision, Singapore-MIT Alliance for Research and Technology, Singapore
| | - Gregory Stephanopoulos
- Disruptive & Sustainable Technologies for Agricultural Precision, Singapore-MIT Alliance for Research and Technology, Singapore.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kang Zhou
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore.,Disruptive & Sustainable Technologies for Agricultural Precision, Singapore-MIT Alliance for Research and Technology, Singapore
| |
Collapse
|
35
|
Network Pharmacology-Based Identification of Potential Targets of Lonicerae japonicae Flos Acting on Anti-Inflammatory Effects. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5507003. [PMID: 34595237 PMCID: PMC8478540 DOI: 10.1155/2021/5507003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/10/2021] [Accepted: 08/25/2021] [Indexed: 12/31/2022]
Abstract
Lonicerae japonicae flos (LJF) is widely used for the treatment of inflammation-related diseases in traditional Chinese medicine (TCM). To clarify the anti-inflammatory mechanism of LJF, 29 compounds with high content in LJF were selected for network pharmacology. Then, a comprehensive network pharmacology strategy was implemented, which involved compound-inflammation-target construction, protein-protein interaction (PPI) network analysis, and enrichment analysis. Finally, molecular docking and in vitro experiments were performed to verify the anti-inflammatory activity and targets of the key compound. As a result, 279 inflammation-associated proteins were identified, which are mainly involved in the AGE/RAGE signaling pathway in diabetic complications, the HIF-1 signaling pathway, the PI3K-AKT signaling pathway, and EGFR tyrosine kinase inhibitor resistance. A total of 12 compounds were linked to more than 35 targets, including apigenin, kaempferol, quercetin, luteolin, and ferulic acid. The results of molecular docking showed that AKT has the most binding activity, exhibiting certain binding activity with 10 compounds, including vanillic acid, protocatechuic acid, secologanic acid, quercetin, and luteolin; the results of qRT-PCR and WB confirmed that two key compounds, secologanic acid and luteolin, could significantly decrease the secretion of TNF-α and the AKT expression of RAW264.7 murine macrophages stimulated by LPS (lipopolysaccharide). These results demonstrate that the comprehensive strategy can serve as a universal method to illustrate the anti-inflammatory mechanisms of traditional Chinese medicine by identifying the pathways or targets.
Collapse
|
36
|
Zhou J, Peng Z, Wang J. Trelagliptin Alleviates Lipopolysaccharide (LPS)-Induced Inflammation and Oxidative Stress in Acute Lung Injury Mice. Inflammation 2021; 44:1507-1517. [PMID: 33751359 DOI: 10.1007/s10753-021-01435-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 01/21/2021] [Accepted: 02/08/2021] [Indexed: 12/14/2022]
Abstract
Acute lung injury (ALI) is an urgent disease lacking effective therapies, resulting in relatively high morbidity and mortality. The pathological mechanism of ALI is reported to be related to excessive inflammation and activated oxidative stress. The present study aims to investigate the protective effects of the DPP-4 inhibitor Trelagliptin against lipopolysaccharide (LPS)-induced ALI and the underlying mechanism. LPS was used to induce ALI mice models. The pathological condition of ALI mice was evaluated using MPO activity assay, lung wet to dry weight ratio detection, and HE staining on the lung tissues. Lung function was assessed using a spirometer. The oxidative stress level in the lung tissues was checked by MDA measurement and GPx detection using commercial kits. The leukocyte and neutrophil numbers were determined using a hemocytometer and the total concentration of protein in the BALF was detected using a bicinchoninic acid method. The expression levels of TNF-α, IL-6, and CXCL2 in the lung tissues were evaluated using qRT-PCR and ELISA. Western blot analysis was used to determine the expression levels of TLR4 and p-NF-κB p65. LPS-induced elevated MPO activity, pulmonary wet to dry weight ratio, airway resistance (RAW), the total number of leukocytes and neutrophils, production of inflammatory factors, decreased pulmonary dynamic compliance (Cdyn), and peak expiratory flow (PEF), and an aggravated histopathological state (such as disordered alveolar structure, significant pulmonary interstitial edema, and large numbers of red blood cells and inflammatory cells in the alveolar cavity) were significantly reversed by the administration of Trelagliptin. The TLR4/NF-κB signaling pathway was activated and oxidative stress was induced by stimulation with LPS; however, both effects were suppressed by the administration of Trelagliptin. Trelagliptin might alleviate LPS-induced inflammation and oxidative stress in acute lung injury mice.
Collapse
Affiliation(s)
- Jia Zhou
- Department of Emergency, The First Affiliated Hospital of University of South China, No.69, Chuanshan Road, Shigu District, Hengyang, 421000, Hunan, China
| | - Zhengliang Peng
- Department of Emergency, The First Affiliated Hospital of University of South China, No.69, Chuanshan Road, Shigu District, Hengyang, 421000, Hunan, China
| | - Jian Wang
- Department of Emergency, The First Affiliated Hospital of University of South China, No.69, Chuanshan Road, Shigu District, Hengyang, 421000, Hunan, China.
| |
Collapse
|
37
|
Herbal Active Ingredients: Potential for the Prevention and Treatment of Acute Lung Injury. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5543185. [PMID: 34258266 PMCID: PMC8245226 DOI: 10.1155/2021/5543185] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023]
Abstract
Acute lung injury (ALI) is a life-threatening clinical syndrome with high morbidity and mortality. The main pathological features of ALI are increased alveolar-capillary membrane permeability, edema, uncontrolled migration of neutrophils to the lungs, and diffuse alveolar damage, resulting in acute hypoxemic respiratory failure. Glucocorticoids, aspirin, and other anti-inflammatory drugs are commonly used to treat ALI. Respiratory supports, such as a ventilator, are used to alleviate hypoxemia. Many treatment methods are available, but they cannot significantly ameliorate the quality of life of patients with ALI and reduce mortality rates. Herbal active ingredients, such as flavonoids, terpenoids, saponins, alkaloids, and quinonoids, exhibit advantages for ALI prevention and treatment, but the underlying mechanism needs further study. This paper summarizes the role of herbal active ingredients in anti-ALI therapy and progresses in the understanding of their mechanisms. The work also provides some references and insights for the discovery and development of novel drugs for ALI prevention and treatment.
Collapse
|
38
|
Wang H, Wu C, Kong D. miR-140-5p Overexpression Protects Against Lipopolysaccharide-Induced Necrotizing Pneumonia via Targeting Toll-Like Receptor 4. Cell Mol Bioeng 2021; 14:339-348. [PMID: 34295443 DOI: 10.1007/s12195-021-00673-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 04/27/2021] [Indexed: 01/04/2023] Open
Abstract
Objective This study is to identify the effects of miRNA-140-5p on necrotizing pneumonia (NP) and its underlying mechanism. Methods The mRNA levels of miRNA-140-5p and TLR4 and secretion of IL-1β, IL-6, and TNF-α in peripheral blood from children with NP and healthy volunteers were determined using qRT-PCR and specific ELISAs. The interactions between miRNA-140-5p and TLR4 were investigated using a dual-luciferase reporter system. Cell viabilities were determined using a CCK-8 assay. qRT-PCR, western blotting, and specific ELISAs were applied to determine the expressions of genes in the cells. Peripheral blood from children with NP had significantly elevated levels of TLR4 but significantly lower levels of miR-140-5p compared to the control. Results Spearman's rank correlation analysis showed a negative correlation between TLR4 and miR-140-5p. miR-140-5p regulated the expressions of TLR4 in A549 cells. Additionally, LPS induced a significant enhancement in the levels of TLR4 but significant reduction in the levels of miR-140-5p. The overexpression of miR-140-5p suppressed cell apoptosis and induced the release of inflammatory cytokines in the LPS-induced A549 cells. Conclusion The underlying mechanisms of miR-140-5p on the regulation of TLR4 are in part by the regulation of p65. The miR-140-5p inhibits necrotizing pneumonia by regulating TLR-4 via TNF-p65 signaling pathway. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-021-00673-0.
Collapse
Affiliation(s)
- Haichao Wang
- School of Biology and Food Engineering, Suzhou University, No. 49 Bianhe Middle Road, Suzhou, 234000 Anhui China
| | - Changhao Wu
- School of Biology and Food Engineering, Suzhou University, No. 49 Bianhe Middle Road, Suzhou, 234000 Anhui China
| | - Dehui Kong
- School of Biology and Food Engineering, Suzhou University, No. 49 Bianhe Middle Road, Suzhou, 234000 Anhui China
| |
Collapse
|
39
|
Luo Q, Zhu J, Zhang Q, Xie J, Yi C, Li T. MicroRNA-486-5p Promotes Acute Lung Injury via Inducing Inflammation and Apoptosis by Targeting OTUD7B. Inflammation 2021; 43:975-984. [PMID: 31940107 DOI: 10.1007/s10753-020-01183-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aim of this article is to study the effect of miR-486-5p in acute lung injury (ALI). MiR-486-5p expression in peripheral blood was determined in ALI patients and healthy volunteers by qRT-PCR. ALI mouse model were reproduced by LPS treatment, and miR-486-5p NC and miRNA-486 inhibitors were injected through trachea. ALI patients' peripheral blood and LPS-induced acute lung injury in mice had significantly higher miR-486-5p levels than control subjects. Inhibition of miR-486-5p by injection with antagomiR-486-5p markedly reduced LPS-induced lung inflammation. Moreover, knockdown of miR-486-5p can reduce protects A549 cell against LPS-induced injury and its corresponding inflammatory response. In addition, Mechanistic analysis indicated that miR-486-5p on the occurrence of ALI is related to the inhibition of OTUD7B activity, which induces the downregulation of inflammatory in ALI. Our results identified miR-486-5p independently associated with ALI. miR-486-5p can mediate the formation of ALI by promoting inflammation.
Collapse
Affiliation(s)
- Qiang Luo
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Zhu
- Department of Anesthesiology, The Fourth Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Zhang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Xie
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengla Yi
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyu Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
40
|
El-Shahat RA, El-Demerdash RS, El Sherbini ES, Saad EA. HCl-induced acute lung injury: a study of the curative role of mesenchymal stem/stromal cells and cobalt protoporphyrin. J Genet Eng Biotechnol 2021; 19:41. [PMID: 33721136 PMCID: PMC7958097 DOI: 10.1186/s43141-021-00139-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 03/02/2021] [Indexed: 02/07/2023]
Abstract
Background This study was designed to investigate bone marrow mesenchymal stem/stromal cells (BM-MSCs) and cobalt protoporphyrin (CoPP) curable effects on HCl-induced acute lung injury (ALI) and its underlying mechanisms hoping this might aid to offer a therapeutic opportunity for ALI. Results Forty male Sprague Dawley rats were randomly allocated into four groups; normal (normal rats), ALI (rats injected with 2 ml hydrochloric acid (HCl)/kg via trachea), ALI + BM-MSCs (ALI rats intravenously injected twice with 1 × 106 BM-MSCs/rat/week), and ALI + CoPP (ALI rats intraperitoneally injected twice with CoPP (0.5 mg/100 g/week)). White blood cells (WBCs), red blood cells (RBCs), hemoglobin (Hb), serum tumor necrosis factor-alpha (TNF-α), lung histopathology, apoptosis markers (caspase-3 and Bcl2), and oxidative stress markers (malondialdehyde (MDA), superoxide dismutase (SOD), and catalase (CAT)) were measured. ALI caused increases in WBCs, TNF-α, caspase-3, and MDA, and morphological damage score of lungs with decreases in RBCs, Hb, Bcl2, SOD, and CAT (p < 0.05). BM-MSCs or CoPP treatment reversed these ALI-induced changes (p < 0.05) towards normal. Conclusions BM-MSCs and CoPP could attenuate ALI by modulation of inflammation, oxidative stress, and apoptosis. Curative roles of BM-MSCs were more effective than those of CoPP. This highlights BM-MSCs as a potent therapy for HCl-associated ALI.
Collapse
Affiliation(s)
- Reham A El-Shahat
- Chemistry Department, Faculty of Science, Damietta University, Damietta, Egypt
| | - Reda S El-Demerdash
- Urology & Nephrology Center, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - El Said El Sherbini
- Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Entsar A Saad
- Chemistry Department, Faculty of Science, Damietta University, Damietta, Egypt.
| |
Collapse
|
41
|
Anemoside B4 Protects against Acute Lung Injury by Attenuating Inflammation through Blocking NLRP3 Inflammasome Activation and TLR4 Dimerization. J Immunol Res 2020; 2020:7502301. [PMID: 33344657 PMCID: PMC7732379 DOI: 10.1155/2020/7502301] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/25/2022] Open
Abstract
Acute lung injury (ALI) is an acute inflammatory process in the lung parenchyma. Anemoside B4 (B4) was isolated from Pulsatilla, a plant-based drug against inflammation and commonly applied in traditional Chinese medicine. However, the anti-inflammatory effect and the mechanisms of B4 are not clear. In this study, we explored the potential mechanisms and anti-inflammatory activity of B4 both in vitro and in vivo. The results indicated that B4 suppressed the expression of iNOS, COX-2, NLRP3, caspase-1, and IL-1β. The ELISA assay results showed that B4 significantly restrained the release of inflammatory cytokines like TNF-α, IL-6, and IL-1β in macrophage cells. In addition, B4 rescued mitochondrial membrane potential (MMP) loss in (lipopolysaccharide) LPS plus ATP stimulated macrophage cells. Co-IP and molecular docking results illustrated that B4 disrupted the dimerization of TLR4. For in vivo results, B4 exhibited a protective effect on LPS and bleomycin- (BLM-) induced ALI in mice through suppressing the lesions of lung tissues, the release of inflammatory cytokines, and the levels of white blood cells, neutrophils, and lymphoid cells in the blood. Collectively, B4 has a protective effect on ALI via blocking TLR4 dimerization and NLRP3 inflammasome activation, suggesting that B4 is a potential agent for the treatment of ALI.
Collapse
|
42
|
Majnooni MB, Fakhri S, Shokoohinia Y, Kiyani N, Stage K, Mohammadi P, Gravandi MM, Farzaei MH, Echeverría J. Phytochemicals: Potential Therapeutic Interventions Against Coronavirus-Associated Lung Injury. Front Pharmacol 2020; 11:588467. [PMID: 33658931 PMCID: PMC7919380 DOI: 10.3389/fphar.2020.588467] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022] Open
Abstract
Since the outbreak of coronavirus disease 2019 (COVID-19) in December 2019, millions of people have been infected and died worldwide. However, no drug has been approved for the treatment of this disease and its complications, which urges the need for finding novel therapeutic agents to combat. Among the complications due to COVID-19, lung injury has attained special attention. Besides, phytochemicals have shown prominent anti-inflammatory effects and thus possess significant effects in reducing lung injury caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Also, the prevailing evidence reveales the antiviral effects of those phytochemicals, including anti-SARS-CoV activity, which could pave the road in providing suitable lead compounds in the treatment of COVID-19. In the present study, candidate phytochemicals and related mechanisms of action have been shown in the treatment/protection of lung injuries induced by various methods. In terms of pharmacological mechanism, phytochemicals have shown potential inhibitory effects on inflammatory and oxidative pathways/mediators, involved in the pathogenesis of lung injury during COVID-19 infection. Also, a brief overview of phytochemicals with anti-SARS-CoV-2 compounds has been presented.
Collapse
Affiliation(s)
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Yalda Shokoohinia
- Pharmaceutical Sciences Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Ric Scalzo Botanical Research Institute, Southwest College of Naturopathic Medicine, Tempe, AZ, United States
| | - Narges Kiyani
- Ric Scalzo Botanical Research Institute, Southwest College of Naturopathic Medicine, Tempe, AZ, United States
| | - Katrina Stage
- Ric Scalzo Botanical Research Institute, Southwest College of Naturopathic Medicine, Tempe, AZ, United States
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Javier Echeverría
- Departamento De Ciencias Del Ambiente, Facultad De Química y Biología, Universidad De Santiago De Chile, Santiago, Chile
| |
Collapse
|
43
|
The odorant receptor OR2W3 on airway smooth muscle evokes bronchodilation via a cooperative chemosensory tradeoff between TMEM16A and CFTR. Proc Natl Acad Sci U S A 2020; 117:28485-28495. [PMID: 33097666 PMCID: PMC7668088 DOI: 10.1073/pnas.2003111117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Odorant sensing GPCRs are the largest gene family in the human genome. We previously found multiple olfactory receptors and their obligate downstream effectors expressed in the smooth muscle of human bronchi. However, the extent to which odorant-sensing receptors (and the ligands to which they respond) on airway smooth muscle (ASM) are physiologically relevant is not established. Here we show that a monoterpene nerol activates the odorant receptor OR2W3 to relax ASM in both cell and tissue models. Surprisingly, the mechanism of action of OR2W3-mediated ASM relaxation involves paradoxical increases in [Ca2+]i that invoke a cooperative activation of TMEM16A and CFTR to compartmentalize calcium and regulate excitation-contraction coupling in human ASM cells. The recent discovery of sensory (tastant and odorant) G protein-coupled receptors on the smooth muscle of human bronchi suggests unappreciated therapeutic targets in the management of obstructive lung diseases. Here we have characterized the effects of a wide range of volatile odorants on the contractile state of airway smooth muscle (ASM) and uncovered a complex mechanism of odorant-evoked signaling properties that regulate excitation-contraction (E-C) coupling in human ASM cells. Initial studies established multiple odorous molecules capable of increasing intracellular calcium ([Ca2+]i) in ASM cells, some of which were (paradoxically) associated with ASM relaxation. Subsequent studies showed a terpenoid molecule (nerol)-stimulated OR2W3 caused increases in [Ca2+]i and relaxation of ASM cells. Of note, OR2W3-evoked [Ca2+]i mobilization and ASM relaxation required Ca2+ flux through the store-operated calcium entry (SOCE) pathway and accompanied plasma membrane depolarization. This chemosensory odorant receptor response was not mediated by adenylyl cyclase (AC)/cyclic nucleotide-gated (CNG) channels or by protein kinase A (PKA) activity. Instead, ASM olfactory responses to the monoterpene nerol were predominated by the activity of Ca2+-activated chloride channels (TMEM16A), including the cystic fibrosis transmembrane conductance regulator (CFTR) expressed on endo(sarco)plasmic reticulum. These findings demonstrate compartmentalization of Ca2+ signals dictates the odorant receptor OR2W3-induced ASM relaxation and identify a previously unrecognized E-C coupling mechanism that could be exploited in the development of therapeutics to treat obstructive lung diseases.
Collapse
|
44
|
Echinacea polysaccharide alleviates LPS-induced lung injury via inhibiting inflammation, apoptosis and activation of the TLR4/NF-κB signal pathway. Int Immunopharmacol 2020; 88:106974. [PMID: 33182056 DOI: 10.1016/j.intimp.2020.106974] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 08/24/2020] [Accepted: 09/01/2020] [Indexed: 11/23/2022]
Abstract
Lung injury is a common critical life-threatening syndrome. Inflammation is a key factor in the pathogenesis of lung injury. It is reported that Echinacea Polysaccharides (EP) has anti-inflammatory activity. However, the effect of EP on lung injury remains unclear. In our study, murine model of lung injury was induced with 2.5 mg/kg LPS before administration of 5 mg/kg or 10 mg/kg EP. EP ameliorated LPS-induced lung pathological damage, along with reduction in lung wet/dry weight ratio and myeloperoxidase activity. EP decreased the number of leukocytes, eosinophils, neutrophils, lymphocytes and macrophages in bronchoalveolar lavage fluid, and the release of tumour necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interleukin-1β (IL-1β) in LPS-treated lung. EP suppressed LPS-induced apoptosis along with down-regulation of Bcl2-associated X (Bax) and cleaved caspase-3 (CC3), and elevated B-cell lymphoma-2 (Bcl-2). Besides, RAW 264.7 cells were treated with EP 100 μg/ml for 1 h and then incubated with 1 μg/ml LPS for 24 h. TNF-α, IL-6 and IL-1β levels were lowered by treatment of EP in LPS-treated RAW 264.7 cells. Moreover, EP down-regulated the expression of toll-like receptor 4 (TLR4), myeloid differentiating factor 88 (MyD88), p-IκBα, nuclear factor kappa-B (NF-κB), p-NF-κB, and up-regulated the inhibitor of NF-κB (IκBα) in vivo and in vitro following LPS induction, which is consistent with the effect of TAK-242. In conclusion, EP may alleviate LPS-induced lung injury via inhibiting inflammation, apoptosis and activation of TLR4/NF-κB signal pathway.
Collapse
|
45
|
Csikós E, Csekő K, Ashraf AR, Kemény Á, Kereskai L, Kocsis B, Böszörményi A, Helyes Z, Horváth G. Effects of Thymus vulgaris L., Cinnamomum verum J.Presl and Cymbopogon nardus (L.) Rendle Essential Oils in the Endotoxin-induced Acute Airway Inflammation Mouse Model. Molecules 2020; 25:molecules25153553. [PMID: 32759721 PMCID: PMC7436258 DOI: 10.3390/molecules25153553] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 01/02/2023] Open
Abstract
Thyme (TO), cinnamon (CO), and Ceylon type lemongrass (LO) essential oils (EOs) are commonly used for inhalation. However, their effects and mechanisms on inflammatory processes are not well-documented, and the number of in vivo data that would be important to determine their potential benefits or risks is low. Therefore, we analyzed the chemical composition and investigated the activity of TO, CO, and LO on airway functions and inflammatory parameters in an acute pneumonitis mouse model. The components of commercially available EOs were measured by gas chromatography-mass spectrometry. Airway inflammation was induced by intratracheal endotoxin administration in mice. EOs were inhaled during the experiments. Airway function and hyperresponsiveness were determined by unrestrained whole-body plethysmography on conscious animals. Myeloperoxidase (MPO) activity was measured by spectrophotometry from lung tissue homogenates, from which semiquantitative histopathological scores were assessed. The main components of TO, CO, and LO were thymol, cinnamaldehyde, and citronellal, respectively. We provide here the first evidence that TO and CO reduce inflammatory airway hyperresponsiveness and certain cellular inflammatory parameters, so they can potentially be considered as adjuvant treatments in respiratory inflammatory conditions. In contrast, Ceylon type LO inhalation might have an irritant effect (e.g., increased airway hyperresponsiveness and MPO activity) on the inflamed airways, and therefore should be avoided.
Collapse
Affiliation(s)
- Eszter Csikós
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pécs, H-7624 Pécs, Hungary; (E.C.); (A.R.A.)
| | - Kata Csekő
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624 Pécs, Hungary; (K.C.); (Á.K.); (Z.H.)
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
| | - Amir Reza Ashraf
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pécs, H-7624 Pécs, Hungary; (E.C.); (A.R.A.)
| | - Ágnes Kemény
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624 Pécs, Hungary; (K.C.); (Á.K.); (Z.H.)
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - László Kereskai
- Department of Pathology, Medical School, University of Pécs, H-7624 Pécs, Hungary;
| | - Béla Kocsis
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, H-7624 Pécs, Hungary;
| | - Andrea Böszörményi
- Department of Pharmacognosy, Faculty of Pharmacy, Semmelweis University, H-1085 Budapest, Hungary;
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624 Pécs, Hungary; (K.C.); (Á.K.); (Z.H.)
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
- PharmInVivo Ltd., H-7629 Pécs, Hungary
| | - Györgyi Horváth
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pécs, H-7624 Pécs, Hungary; (E.C.); (A.R.A.)
- Correspondence: ; Tel.: +36-72-503650-28823
| |
Collapse
|
46
|
Yang Y, Ding Z, Wang Y, Zhong R, Feng Y, Xia T, Xie Y, Yang B, Sun X, Shu Z. Systems pharmacology reveals the mechanism of activity of Physalis alkekengi L. var. franchetii against lipopolysaccharide-induced acute lung injury. J Cell Mol Med 2020; 24:5039-5056. [PMID: 32220053 PMCID: PMC7205831 DOI: 10.1111/jcmm.15126] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/03/2020] [Accepted: 01/21/2020] [Indexed: 12/11/2022] Open
Abstract
Acute lung injury (ALI) is an important cause of mortality of patients with sepsis, shock, trauma, pneumonia, multiple transfusions and pancreatitis. Physalis alkekengi L. var. franchetii (Mast.) Makino (PAF) has been extensively used in Chinese folk medicine because of a good therapeutic effect in respiratory diseases. Here, an integrated approach combining network pharmacology, proton nuclear magnetic resonance-based metabolomics, histopathological analysis and biochemical assays was used to elucidate the mechanism of PAF against ALI induced by lipopolysaccharide (LPS) in a mouse model. We found that the compounds present in PAF interact with 32 targets to effectively improve the damage in the lung undergoing ALI. We predicted the putative signalling pathway involved by using the network pharmacology and then used the orthogonal signal correction partial least-squares discriminant analysis to analyse the disturbances in the serum metabolome in mouse. We also used ELISA, RT-qPCR, Western blotting, immunohistochemistry and TUNEL assay to confirm the potential signalling pathways involved. We found that PAF reduced the release of cytokines, such as TNF-α, and the accumulation of oxidation products; decreased the levels of NF-κB, p-p38, ERK, JNK, p53, caspase-3 and COX-2; and enhanced the translocation of Nrf2 from the cytoplasm to the nucleus. Collectively, PAF significantly reduced oxidative stress injury and inflammation, at the same time correcting the energy metabolism imbalance caused by ALI, increasing the amount of antioxidant-related metabolites and reducing the apoptosis of lung cells. These observations suggest that PAF may be an effective candidate preparation alleviating ALI.
Collapse
Affiliation(s)
- Yanni Yang
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zihe Ding
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yi Wang
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Renxing Zhong
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanlin Feng
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Tianyi Xia
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuanyuan Xie
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Bingyou Yang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zunpeng Shu
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
47
|
Ji X, Guo Y, Qiu Q, Wang Z, Wang Y, Ji J, Sun Q, Cai Y, Zhou G. [Molecular mechanism underlying the inhibitory effect of propofol on lipopolysaccharide-induced pyroptosis of mouse bone marrow-derived macrophages]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:525-530. [PMID: 32895145 DOI: 10.12122/j.issn.1673-4254.2020.04.12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the molecular mechanism underlying the inhibitory effect of propofol on pyroptosis of macrophages. METHODS Macrophages derived from bone marrow were extracted and divided into three groups: control group, LPS+ATP group and propofol+LPS+ATP group. The control group was not given any treatment; LPS+ATP group was given LPS 1 μg/mL stimulation for 4 h, then ATP 4 mM stimulation for 1 h; Propofol+LPS+ATP group was given propofol+LPS 1 μg/mL stimulation for 4 h, then ATP stimulation for 1 h. After treatment, the supernatant and cells of cell culture were collected. the cell activity was detected by CCK8 and flow cytometry. The inflammatory cytokines IL-1βand IL-18 were detected by Elisa. Western blot was used to detect the expression of caspase-1 protein and TLR4 on cell membran Immunohistochemical fluorescence was used to detect apoptosis of cells. RESULTS LPS+ATP significantly decreased the viability of the macrophages and increased the cellular production of IL-1β and IL-18, activation of caspase-1 protein and the expression of TLR-4 on the cell membrane (P < 0.05). Treatment with propofol obviously reversed the changes induced by LPS+ATP. CONCLUSIONS LPS+ATP can induce pyroptosis of mouse bone marrow-derived macrophages, and propofol effectively inhibits such cell death, suggesting that propofol anesthesia is beneficial during operation and helps to regulate the immune function of in patients with sepsis.
Collapse
Affiliation(s)
- Xuexia Ji
- Department of Anesthesiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yuanbo Guo
- Department of Anesthesiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Qianqi Qiu
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou 510623, China
| | - Zhipeng Wang
- Department of Anesthesiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yan Wang
- Department of Science and Education, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jinquan Ji
- Department of Anesthesiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Qiang Sun
- Department of Anesthesiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yujing Cai
- Department of Anesthesiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Guobin Zhou
- Department of Anesthesiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| |
Collapse
|
48
|
Lee SY, Kim MJ, Jang S, Lee GE, Hwang SY, Kwon Y, Hong JY, Sohn MH, Park SY, Yoon HG. Plumbagin Suppresses Pulmonary Fibrosis via Inhibition of p300 Histone Acetyltransferase Activity. J Med Food 2020; 23:633-640. [PMID: 32311286 DOI: 10.1089/jmf.2019.4670] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic fibrosing interstitial lung disease with a poor prognosis similar to that of malignancy. The causes of IPF are not clearly known, and there is no effective therapy to date. In this study, the natural compound plumbagin, which was isolated from Plumbago rosea root extract, was screened for p300 inhibitory activity. Plumbagin specifically inhibited the activity of p300 toward histone acetyltransferases. Plumbagin treatment significantly suppressed transforming growth factor-β-induced profibrotic target-gene expression and proliferation of fibroblast cell lines. Moreover, plumbagin significantly inhibited bleomycin-induced pulmonary fibrosis in mice. Taken together, these data demonstrate the inhibitory effects of plumbagin on lung fibrosis and its promise as a therapeutic agent for IPF.
Collapse
Affiliation(s)
- Soo Yeon Lee
- Brain Korea 21 PLUS Project for Medical Sciences, Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Mi Jeong Kim
- Brain Korea 21 PLUS Project for Medical Sciences, Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Subhin Jang
- Brain Korea 21 PLUS Project for Medical Sciences, Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Gyeong-Eun Lee
- Brain Korea 21 PLUS Project for Medical Sciences, Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Soo-Yeon Hwang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
| | - Youngjoo Kwon
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
| | - Jung Yeon Hong
- Brain Korea 21 PLUS Project for Medical Sciences, Department of Pediatrics and Institute of Allergy, Severance Medical Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Myung Hyun Sohn
- Brain Korea 21 PLUS Project for Medical Sciences, Department of Pediatrics and Institute of Allergy, Severance Medical Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Soo-Yeon Park
- Brain Korea 21 PLUS Project for Medical Sciences, Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ho-Geun Yoon
- Brain Korea 21 PLUS Project for Medical Sciences, Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
49
|
Boullerne AI, Adami GR, Schwartz JL, Skias D, Maienschein-Cline M, Green SJ, Feinstein DL. Deep DNA metagenomic sequencing reveals oral microbiome divergence between monozygotic twins discordant for multiple sclerosis severity. J Neuroimmunol 2020; 343:577237. [PMID: 32289594 DOI: 10.1016/j.jneuroim.2020.577237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/06/2020] [Accepted: 04/06/2020] [Indexed: 12/17/2022]
Abstract
In contrast to gut, the oral microbiome of MS patients has not been characterized. Deep sequencing of saliva DNA from a pair of monozygotic twins (MSF1 with relapsing remitting MS; MSF2 with clinically isolated syndrome) identified 2036 bacterial species. Relative abundances of 3 phyla were higher, and 3 lower in MSF1 versus MSF2. Species diversity was greater in MSF2, and 20 abundant species differed at least 2-fold. Pathway analysis identified 116 functional hierarchies differing 50% or more. Although limited to one pair of twins, our data suggests that oral microbiome analysis may be useful for diagnosis or monitoring therapeutic efficacy.
Collapse
Affiliation(s)
- Anne I Boullerne
- Department of Anesthesiology, University of Illinois, Chicago, IL, USA.
| | - Guy R Adami
- Department of Oral Medicine and Diagnostic Services, University of Illinois, Chicago, IL, USA
| | - Joel L Schwartz
- Department of Oral Medicine and Diagnostic Services, University of Illinois, Chicago, IL, USA
| | - Demetrios Skias
- Department of Neurology and Rehabilitation, University of Illinois, Chicago, IL, USA
| | | | - Stefan J Green
- Research Resources Center, University of Illinois, Chicago, IL, USA
| | - Douglas L Feinstein
- Department of Anesthesiology, University of Illinois, Chicago, IL, USA; Jesse Brown VA Medical Center, Chicago, IL, USA.
| |
Collapse
|
50
|
Liu P, Feng Y, Li H, Chen X, Wang G, Xu S, Li Y, Zhao L. Ferrostatin-1 alleviates lipopolysaccharide-induced acute lung injury via inhibiting ferroptosis. Cell Mol Biol Lett 2020; 25:10. [PMID: 32161620 PMCID: PMC7045739 DOI: 10.1186/s11658-020-00205-0] [Citation(s) in RCA: 382] [Impact Index Per Article: 76.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/17/2020] [Indexed: 02/07/2023] Open
Abstract
Background Ferroptosis is a newly recognized type of cell death, which is different from traditional necrosis, apoptosis or autophagic cell death. However, the position of ferroptosis in lipopolysaccharide (LPS)-induced acute lung injury (ALI) has not been explored intensively so far. In this study, we mainly analyzed the relationship between ferroptosis and LPS-induced ALI. Methods In this study, a human bronchial epithelial cell line, BEAS-2B, was treated with LPS and ferrostatin-1 (Fer-1, ferroptosis inhibitor). The cell viability was measured using CCK-8. Additionally, the levels of malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), and iron, as well as the protein level of SLC7A11 and GPX4, were measured in different groups. To further confirm the in vitro results, an ALI model was induced by LPS in mice, and the therapeutic action of Fer-1 and ferroptosis level in lung tissues were evaluated. Results The cell viability of BEAS-2B was down-regulated by LPS treatment, together with the ferroptosis markers SLC7A11 and GPX4, while the levels of MDA, 4-HNE and total iron were increased by LPS treatment in a dose-dependent manner, which could be rescued by Fer-1. The results of the in vivo experiment also indicated that Fer-1 exerted therapeutic action against LPS-induced ALI, and down-regulated the ferroptosis level in lung tissues. Conclusions Our study indicated that ferroptosis has an important role in the progression of LPS-induced ALI, and ferroptosis may become a novel target in the treatment of ALI patients.
Collapse
Affiliation(s)
- Pengfei Liu
- 1Department of Anesthesiology, The 2nd Clinical Medical College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, 518020 China.,2Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632 China
| | - Yetong Feng
- 3Health Science Center, School of Basic Medical Sciences, Shenzhen University, Shenzhen, 518037 China
| | - Hanwei Li
- 1Department of Anesthesiology, The 2nd Clinical Medical College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, 518020 China.,4Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, 510280 China
| | - Xin Chen
- 5Department of Laboratory Medicine, The 2nd Clinical Medicine College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, 518020 China
| | - Guangsuo Wang
- 6Department of Thoracic Surgery, The 2nd Clinical Medicine College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, 518020 China
| | - Shiyuan Xu
- 4Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, 510280 China
| | - Yalan Li
- 7Department of Anesthesiology, First Affiliated Hospital of Jinan University, Guangzhou, 510632 China.,2Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632 China
| | - Lei Zhao
- 1Department of Anesthesiology, The 2nd Clinical Medical College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, 518020 China.,2Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632 China
| |
Collapse
|